cassava starch as modified release excipient in selected

134
Cassava starch as modified release excipient in selected gliclazide oral dosage forms WC du Preez B.Pharm 21638098 Dissertation submitted in fulfilment of the requirements for the degree Magister Scientiae of Pharmaceutics at the Potchefstroom Campus of the North-West University Supervisor: Dr JM Viljoen Co-Supervisor: Prof JH Steenekamp November 2015

Upload: others

Post on 23-Mar-2022

9 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Cassava starch as modified release excipient in selected

Cassava starch as modified release excipient in selected gliclazide oral dosage forms

WC du Preez

B.Pharm

21638098

Dissertation submitted in fulfilment of the requirements for the degree Magister Scientiae of Pharmaceutics at the

Potchefstroom Campus of the North-West University

Supervisor: Dr JM Viljoen

Co-Supervisor: Prof JH Steenekamp

November 2015

Page 2: Cassava starch as modified release excipient in selected

1

Our deepest fear is not that we are inadequate. Our deepest fear is that we

are powerful beyond measure. It is our light, not our darkness, that most

frightens us. We ask ourselves, who am I to be brilliant, gorgeous, talented,

fabulous? Actually, who are you not to be? You are a child of God. Your

playing small doesn't serve the world. There's nothing enlightened about

shrinking so that other people won't feel insecure around you. We are all

meant to shine, as children do. We were born to make manifest the glory of

God that is within us. It's not just in some of us; it's in everyone. And as we

let our own light shine, we unconsciously give other people permission to do

the same. As we're liberated from our own fear, our presence automatically

liberates others.

Marianne Williamson

~||~

Dedicated to those who’ve never stopped praying for me,

my mentors and loved ones.

Page 3: Cassava starch as modified release excipient in selected

2

FOREWORD

A great journey has many steps and obstacles. To attempt such a journey requires, patience,

perseverance, passion and fortitude. Each of these is necessary but they alone cannot sustain

any endeavor as this. Where, these personal strengths falter support, care and guidance from

loved ones and mentors, sustain you through trying times, times when failure seems to be the

inevitable outcome. This in mind, I would like to give my sincerest gratitude to the following:

My Lord and Sheppard, thank You for the guidance I have received in this journey of discovery.

I thank You, for the perseverance, tenacity and fortitude I have been blessed with in order to

attempt and complete this chapter in my life. I thank You, for my faith and the path You have

lead me since the first day I have met You.

My late grandfather, Schalk, and my grandmother, Vonnie, thank you for all your love, pride and

endless prayers.

My parents, Este and Koos, thank you for your sacrifice, support, arguments and love that has

guided me all the way, making this chapter in my life possible.

Tannie Suzette and my brother Werner, thank you for your strength, prayers and faith in me.

All my friends, named and unnamed, you all gave me life in these few years, all the coffee, all

the laughs and all the tears.

A few in particular, Lezaan-marie Erasmus, Trizel du Toit, Lonette Wallis, Carlemi Calitz, Ruan

Joubert, Elizca Pretorius, Jacques Scholtz, Zandré Smith, Gerdus Kruger, Thokozile Okaecwe,

Angelique Lewies, Jaco Wentzel and Alissa Jooste, thank you for the advice, strength and

friendship. This experience could not be attempted or survived without it or without your special

“sanity”.

All my colleagues, fellow postgraduate students and friends in the office, you have made my

days full of life, insight and wisdom. The camaraderie and friendship was vital to complete this

degree.

Dr. Joe Viljoen, my study leader and postgraduate mother, who I might have driven mad at one

point or another; constantly popping in with coffee in hand and a chat, thank you for your faith in

my abilities, the patience with my writing and the firm though tender kindness you have shown

me every day since we’ve met.

Page 4: Cassava starch as modified release excipient in selected

3

Prof. Jan Steenekamp, thank you for making my choice in specialty so easy, if not for your pride

in pharmaceutics, I would certainly not have pursued this field or succeeded in it. Thank you for

all the assistance with the dissolution studies, templates and financial support.

Prof. Sandra van Dyk, thank you for your support, both financially and professionally.

Dr. Louwrens Tiedt, I thank you for your facilities and help with my SEM and micrographs.

Dr. Frans Smith, from pharmaceutical chemistry, thank you for your assistance in understanding

my IR-spectra and helping me with the FTIR-analysis.

Niel Barnard and SPIN®, thank you for all the physical analysis you helped me with.

Anriette Pretorius, the librarian, thank you for all the assistance in finding hard to come by

articles.

Jacques Scholtz and the late, Jaco van der Colff, thank you for input and willingness to help me

with my beads and tablets.

Lizl du Toit and Etienne Marais, thank you for helping me understand and use the

UV-spectrometer.

The “Tannies”, Dr. Maides Malan and Mrs. Mariette Fourie, thank for all your kindness, wisdom

and guidance you have given me.

Liezl (Lee) Badenhorst, thank you for all the social events and the opportunity to be your demi.

Pharmacen® including its members, associates and facilities, for affording me the opportunity to

conduct and complete a postgraduate degree.

These are just a few words of gratitude, they cannot convey the full extent and breadth of my

feelings in regard, to each participant and individual named, unnamed, known and unknown.

Page 5: Cassava starch as modified release excipient in selected

4

TABLE OF CONTENTS

Foreword .................................................................................. 2

Table of Contents .................................................................... 4

1.1 Aim .................................................................................. 16

1.2 Background .................................................................... 16

1.3 Objectives ....................................................................... 19

2.1 Introduction .................................................................... 20

2.1.1 Treatment of type 2-diabetes ...................................................... 22

2.1.1.1 Gliclazide ..............................................................................................................23

2.2 Solid Oral Dosage Forms ................................................ 25

2.2.1 Formulation of solid oral dosage forms ....................................... 25

2.2.1.1 Excipients used in formulations ............................................................................26

2.2.2 Manufacturing methods of solid oral dosage forms ................... 28

2.2.2.1 Wet granulation ....................................................................................................28

2.2.2.2 Dry granulation .....................................................................................................29

2.2.2.3 Direct compression ...............................................................................................29

2.2.2.4 Extrusion-Spheronised pharmaceutical pellets .....................................................29

2.3 Immediate release compared to modified release solid

oral dosage forms .......................................................... 30

2.3.1 Types of immediate release solid oral dosage forms ................. 31

2.3.1.1 Conventional release solid oral dosage forms ......................................................31

2.3.1.2 Effervescent Tablets .............................................................................................31

2.3.1.3 Chewable Tablets .................................................................................................32

2.3.1.4 Sublingual and Buccal Tablets..............................................................................32

2.3.1.5 Multi-layer tablets .................................................................................................33

2.3.1.6 Lozenges ..............................................................................................................33

2.3.2 Modified release solid oral dosage forms ................................... 33

2.3.2.1 Coated tablets ......................................................................................................33

Page 6: Cassava starch as modified release excipient in selected

5

2.3.2.2 Diffusion-controlled tablets ...................................................................................34

2.3.2.3 Dissolution-controlled tablets ................................................................................34

2.3.2.4 Erosion controlled tablets .....................................................................................34

2.3.2.6 Osmosis controlled tablets ....................................................................................34

2.3.2.7 Multi-layer tablets .................................................................................................35

2.3.2.8 Multi-particulates ..................................................................................................36

2.3.2.8.1 Layering .............................................................................................................37

2.3.2.8.2 Freeze pelletisation ............................................................................................37

2.3.2.8.3 Cryopellitisation ..................................................................................................38

2.3.2.8.4 Hot-melt extrusion ..............................................................................................38

2.3.2.8.5 Extrusion-spheronisation ....................................................................................38

2.4 Starch as a versatile excipient ...................................... 39

2.4.1 Cassava ......................................................................................... 41

2.5 Summary ......................................................................... 44

3.1 INTRODUCTION .............................................................. 45

3.2 Materials ......................................................................... 45

3.3 Characterisation of cassava starches ........................... 46

3.3.1 Thermoanalytical characterisation ............................................. 47

3.3.1.1 Differential scanning calorimetric (DSC) analysis ..................................................47

3.3.1.2 Thermogravimetric analysis ..................................................................................47

3.3.1.3 Karl-Fischer titration .............................................................................................48

3.3.2 Infrared (IR) analysis .................................................................... 48

3.4 Solid oral dosage forms.................................................. 49

3.4.1 Preparation of beads .................................................................... 49

3.4.1 Morphology of powder particles and bead formulations ............ 50

3.4.1.1 Scanning electron microscopy (SEM) ...................................................................50

3.4.1.2 Particle size analysis ............................................................................................51

3.5 Flow properties .............................................................. 52

3.5.1 Critical orifice diameter ............................................................... 52

Page 7: Cassava starch as modified release excipient in selected

6

3.5.2 Flow rate ....................................................................................... 53

3.5.4 Angle of repose ............................................................................. 54

3.5.4 Powder density ............................................................................. 55

3.5.5 Compressibility ............................................................................. 56

3.6 Evaluation of the bead formulations .............................. 57

3.6.1 Friability ........................................................................................ 57

3.6.2 Swelling and mass loss ................................................................ 57

3.6.3 Disintegration ............................................................................... 58

3.6.4 Ultraviolet-spectrophotometric analysis ..................................... 58

3.6.4.1 Standard curve .....................................................................................................59

3.6.4.1.1 Interday precision ...............................................................................................59

3.6.4.1.2 Intraday precision ...............................................................................................59

3.6.5 Dissolution Behaviour .................................................................. 59

3.6.5.1 Assay ...................................................................................................................60

3.6.5.2 Dissolution studies ................................................................................................60

3.7 Statistical analysis ......................................................... 60

3.8 Summary ......................................................................... 61

4.1 Introduction .................................................................... 63

4.2 Physical characteristics of Cassava starch .................. 64

4.2.1 Moisture content and Thermal analysis ...................................... 64

4.2.2 Infrared-spectroscopy .................................................................. 67

4.3 preliminary experiments and bead manufacturing ....... 68

4.4 Morphology and size ....................................................... 72

4.4.1 Morphology ................................................................................... 72

4.4.2 Size distribution of powder particles .......................................... 75

4.5 Flow properties .............................................................. 76

Page 8: Cassava starch as modified release excipient in selected

7

4.6 Evaluation of bead formulations .................................... 78

4.6.1 Friability ........................................................................................ 79

4.6.2 Swelling and mass loss ................................................................ 80

4.6.3 Disintegration ............................................................................... 82

4.6.4 Dissolution behaviour and statistical analyses .......................... 82

4.6.4.1 Standard curve .....................................................................................................82

4.6.4.2 Linearity ................................................................................................................82

4.6.4.3 Dissolution ............................................................................................................83

4.7 Summary ......................................................................... 86

5.1 SUMMARY & FUTURE PROSPECTS ............................... 88

5.2 FUTURE PROSPECTS ..................................................... 90

References............................................................................. 91

Annexure A .......................................................................... 108

Annexure B .......................................................................... 116

Annexure C .......................................................................... 128

Page 9: Cassava starch as modified release excipient in selected

8

LIST OF FIGURES

Figure 2.1: Chemical structure of gliclazide

Figure 2.2: Arbitrary graph comparing immediate and controlled drug release

Figure 2.3: Sublingual and buccal route of administration

Figure 2.4: Example of an osmotically controlled release tablet

Figure 2.5: Layering process for pharmaceutical beads

Figure 2.6: Radial extruder

Figure 2.7: Molecular and macroscopic structure of amylose and amylopectin

Figure 2.8: Illustration of cassava plant and root

Figure 3.1: Apparatus used for the critical orifice diameter determination

Figure 3.2: Angle of repose of a resting powder heap

Figure 4.1: Average moisture content of donated and the purchased Cassava starch, at

40°C for various drying times

Figure 4.2: Thermograms of donated Cassava starch

Figure 4.3: Thermograms of purchased Cassava starch

Figure 4.4: Overlay of IR-spectra for the donated and purchased starch

Figure 4.5: IR-spectra form FTIR of the donated and purchased Cassava starch

Figure 4.6: Scanning electron micrographs of purchased and donated starch

Figure 4.7: SEM-micrographs of the different bead formulations

Figure 4.8: Size distribution histograms of starches and beads

Figure 4.9: Cumulative mass increase or decrease of Avicel® beads as a function of time

(min) after exposure to calibrated pH environments

Figure 4.10: Standard curve of gliclazide dissolved in 2:3 methanol:HCl solution

Figure 4.11: Percentage of the drug dissolved as a function of time (min) within pH calibrated

medium form simulating either a acidic or alkaline gastric environments

Page 10: Cassava starch as modified release excipient in selected

9

Figure A.I: Example graphs of size distribution graphs for starch powders and bead

formulation

Page 11: Cassava starch as modified release excipient in selected

10

LIST OF TABLES

Table 2.1: Physicochemical properties of gliclazide

Table 2.2: Excipient types and examples

Table 2.3: Content properties of cassava

Table 2.4: Physicochemical properties of cassava variants

Table 3.1: Pharmaceutical materials employed in the various formultaions, batch numbers

and suppliers

Table 3.2: Variables and different levels of each variable as employed in this study.

Table 3.3: Flow quality of powders for various angles of repose

Table 3.4: Flow quality as indicated by Carr’s index and the Hausner ratio

Table 4.1: Identifiers for each bead formulation and the composition of each formulation

Table 4.2: Selected formulations and respective excipients and concentration

Table 4.2: Flow properties of starches and bead formulations

Table 4.3: Percentage friability of bead formulations

Table 4.4: Mean dissolution time and similarity factor values for each bead formulation and

Diamicron®

Table A.I: Karl-Fischer titration values for moisture content of the donated Cassava starch

Table A.II: Karl-Fischer titration values for moisture content of the purchased Cassava

starch

Table A.III: Size distribution values of starch powders and bead formulations

Table B.I: Time and flow rate for Cassava starch powders and bead formulations

Table B.II: Parameters and values relating to angle of repose, the angle of repose and

critical orifice diameter

Table B.III: Volumes, densities and compressibility data

Table B.IV: Swelling and erosion data

Page 12: Cassava starch as modified release excipient in selected

11

Table B.V: Friability parameters and data

Table B.VI: Disintegration times

Table C.I: Linearity and validation data for gliclazide in acidic medium

Table C.II: Linearity and validation data for gliclazide in alkaline medium

Table C.III: Dissolution study data for bead formulations and Diamicron®

Page 13: Cassava starch as modified release excipient in selected

12

Abstract

Cassava starch as modified release excipient in oral dosage

forms using gliclazide as model drug

Solid oral dosage forms are still the most leading delivery system employed commercially due to

the ease in which it can be handled, administered and even transported. Several varieties of

solid oral dosage forms are commercially available which include different types of tablets,

capsules, multi-unit particulate systems as well as medicated lozenges. Different designs and

manufacturing methods are used for solid oral dosage forms resulting in different release

mechanisms. Drug release is an important consideration during dosage form design especially

for drugs with short half-lives. These types of drugs require regularly timed dosing intervals.

More dose intervals can impede the adherence to therapy, because patients might forget a

dose. The lack in adherence adversely affects the treatment protocol necessary for the

management of disease. To overcome adversities and to modify drug release, various methods

can be employed in order to provide a desirable therapeutic product, including alternative

manufacturing methods and the addition of specialised excipients. One of the most promising

manufacturing methods to date regarding modified release, whether sustained, controlled or

multi-dose release, is the production of pharmaceutical pellets, more commonly known as

beads. Several methods can be employed in order to produce beads. For this study it was

opted to use a method, which has extensively been researched since the 1950s known as

extrusion-spheronisation.

Starches and starch based products have been utilised for many years as multifunctional

excipients in the production of solid oral dosage forms. For instance, starches have been used

as fillers, binders and disintegrants. The polymer rich matrix of a starch makes it highly versatile

in these applications. Furthermore, the low cost involved in manufacturing or sourcing starch

and starch based products, also makes it a commercially viable alternative to other market

available excipients which might be more expensive. Cassava is one of the world’s most

predominant sources of starch. It is globally grown and sourced in sub-tropic environments.

Being a sustainable product which produces a high yield of starch, this study investigated the

applicability of cassava starch as a filler in bead formulations using gliclazide as model drug.

Page 14: Cassava starch as modified release excipient in selected

13

Physical characteristics and flowability of cassava starch were evaluated with various methods,

which included thermo-analysis, moisture content, infrared spectrometry, and flow properties.

Beads were evaluated in order to determine whether extrusion-spheronisation improved the flow

of the starch. The physical characteristics such as friability, swelling and erosion, and

disintegration were also evaluated. Dissolution testing and analysis provided profiles which

were assessed and compared to a commercially available product, Diamicron®.

It was evident from the study that cassava is not the ideal filler to include in the manufacture of

beads, even though a single cassava bead formulation did provide prolonged release of the

drug over a 12 h period. Approximately 60% of the drug was pharmaceutically available within

the first 30 min of dissolution assessment and the remaining 40% dissolved slowly over the

remaining duration of the study. The dissolution profile obtained for this particular formulation

correlated with the arbitrary release profile of sustained drug release. It could therefore be

concluded that a product could indeed be produced which may be a viable candidate as a

commercially substitute for the current commercially available product, in terms of cost-

effectiveness and sustainability. From the study it was also evident that Avicel® provided a

better prolonged release profile in terms of mean dissolution time. Avicel® formulations proved

to render the most similar release profiles to that of the reference product, Diamicron®.

Keywords: Cassava, Starch, Extrusion-spheronisation, modified release, solid oral dosage

forms (SODFs), flowability, powder flow. Gliclazide, Avicel®, beads,

microcrystalline cellulose (MCC)

Page 15: Cassava starch as modified release excipient in selected

14

Uittreksel

Kassawestysel as ʼn vrystellings-modifiserende hulpstof in

vaste doseervorms met gliklasied as modelgeneesmiddel

Orale vaste doseervorms is die gewildste geneesmiddelafleweringssisteme wat kommersieel

beskikbaar is. Hierdie gewildheid kan toegeskryf word aan die gemak waarmee dit hanteer,

toegedien , en selfs vervoer word. Verskeie tipes orale vaste doseervorms is kommersieel

beskikbaar insluitend tablette, kapsules, meervoudige partikulêre sisteme en suigtablette.

Verskillende ontwerpe en vervaardigingsmetodes word gebruik in die bereiding van vaste

doseervorms ten einde verskillende tipes geneesmiddelvrystelling te verkry.

Geneesmiddelvrystelling is ʼn uiters belangrike oorweging tydens doseervormontwerp, veral vir

geneesmiddels met kort halfleeftye. Geneesmiddels met kort halfleeftye benodig gereelde

doserings op spesifieke tye. Meervoudige doseerskedules kan tot swak pasiënt-

meewerkendheid lei, wat kan lei tot een of meer oorgeslane dosisse. Swak

pasiëntmeewerkendheid veroorsaak ʼn afname in behandelingseffektiwiteit. Ten einde hierdie

struikelblokke te oorkom en geneesmiddelvrystelling te verbeter, word verskeie metodes

ingespan, onder andere, alternatiewe vervaardigingsmetodes en die gebruik van spesiale

hulpstowwe. ʼn Belowende vervaardigingsmetode wat tans baie navorsingsaandag ontvang om

verbeterde geneesmiddelvrystelling, hetsy dit verlengde, beheerde of meervoudige

dosisvrystelling behels, is die vervaardiging van farmaseutiese korrels, byvoorbeeld krale.

Verskeie vervaardigingsmetodes kan gebruik word om krale te vervaardig. In hierdie studie is

daar gebruik gemaak van ʼn metode wat sedert die 1950s breedvoerig nagevors is, naamlik

uitpers-sferonisering.

Stysels en styselgebaseerde produkte word al vir jare as multifunksionele hulpstowwe in die

vervaardiging van orale vaste doseervorms gebruik. So byvoorbeeld word stysel of stysel-

gebaseerde produkte onder andere as vulstof, bindmiddel en disintegreermiddel aangewend.

Die polimeerryke matriks verleen aan stysel die vermoë om as multifunksionele hulpstof gebruik

te word. Die lae vervaardigingskoste asook maklike verkryging van stysel en styselgebaseerde

produkte maak dit ʼn kommersieel aanvaarde alternatief as plaasvervanger vir duurder

hulpstowwe. Die Kassaweplant is een van die wêreld se mees algemene bronne van stysel.

Dit kom wêreldwyd voor in subtropiese gebiede. Omdat dit ʼn volhoubare bron is, wat ʼn hoë

Page 16: Cassava starch as modified release excipient in selected

15

stysel-opbrengs lewer, is daar in hierdie studie ondersoek ingestel na die bruikbaarheid van

Kassawestysel as vrystellingsmodifiserende hulpstof in die bereiding van krale.

Die fisiese eienskappe en vloeibaarheid van kassawestysel is gekarakteriseer met die gebruik

van verskeie metodes, waaronder termiese analise, voginhoudbepaling en infrarooi-

spektrometrie. Bereide krale is ook geëvalueer in terme van swelling, verbrokkeling,

disintegrasie en dissolusiegedrag.

Die resultate van die studie het getoon dat kassawestysel nie optimale krale gelewer het nie.

Ten spyte hiervan het ʼn enkele kassawe-kraalformulering verlengde vrystelling van gliklasied

oor ʼn 12 h tydperk getoon. Ongeveer 60% van die geneesmiddel is binne die eerste 30 min.

vrygestel, en die oorblywende 40% is in die oorblywende tyd van die studie vrygestel. Die

dissolusieprofiel het ooreengestem met die arbitrêre vrystellingsprofiel vir volhoude

geneesmiddelvrystelling. Vanuit die data kon gesien word dat die moontlikheid bestaan om ʼn

formulering te berei wat oor die potensiaal beskik om huidige kommersieel beskikbare produkte,

in terme van koste-effektiwiteit en volhoubaarheid, te vervang. Avicel® (mikrokristallyne

sellulose), - tans die standaard vir kraalbereiding — is ook in die studie gebruik om as maatstaf

vir kassawestysel te dien. Uit die resultate was dit duidelik dat Avicel® ʼn beter verlengde

vrystellingsprofiel verskaf het in terme van die gemiddelde dissolusie tyd. Avicel®-formulerings

het ook bewys dat die vrystellingsprofiel van die verwysingsproduk, Diamicron®, nageboots kan

word onder spesifieke eksperimentele toestande.

Sleutelwoorde: Kassawestysel, uitpers-sfeervorming, gemodifiseerde vrystelling, vaste orale

doseer vorme, dissolusie studies, vloeibaarheid, poeiervloei, gliklasied,

Avicel®, krale, mikrokristallyne sellulose.

Page 17: Cassava starch as modified release excipient in selected

16

Chapter 1

AIMS AND OBJECTIVES

1.1 AIM

The aim of this study was to investigate the possible application of cassava starch as an

excipient in a modified release solid oral dosage form. In conjunction with this investigation it

was also considered prudent to investigate the effects of using a multi-unit pellet (or

particulate) system as a modified release solid oral dosage form.

1.2 BACKGROUND

Changes in economies and socio-economic diversity as a result of globalisation and the growth

in consumerism have had both advantageous and disadvantageous consequences; e.g.

improved transport infrastructure, communication systems, energy generations, increased

health risks and increased levels of unemployment (Reddy et al., 2006:1-9; Storper 2000:

107-114). This is especially evident on the African continent. An area of concern, however, not

only on the sub-Saharan African continent, but also in developed economies such as Europe,

Japan and Northern America, is lifestyle dependent health risks. Lifestyle dependent health

risks in developed nations are a result of increased consumerism and an ever decreasing labour

intensive lifestyle (Badawi et al., 2004:76), whereas sub-Saharan Africa and other global

counterparts have increased health risks as a result of limited or no access to sufficient

resources, e.g. medical personnel, medical equipment and medication. Consequences of

health risks include sexually transmitted infections, low infant mortality rate, low female health

care and even the escalation in lifestyle dependent health risks. The latter is brought forth not

only by urbanisation but also the impoverishment of economically unstable nations

(Addo et al., 2007:1013; Meyrowitsch et al., 2007:32).

One of the most common lifestyle dependent health concerns is certainly insufficient glycaemic

control (Zimmet et al., 2001:782). Hyper- and hypoglycaemia are two pathological

manifestations of insulin insufficiencies, brought on either by defects in insulin secretion or

desensitised tissue response to insulin and glucose levels. In this study; only hyperglycaemia

was addressed.

Page 18: Cassava starch as modified release excipient in selected

17

The most predominant disease characterised by hyperglycaemia is diabetes mellitus type 1

(DMT-1) and 2 (DMT-2). DMT-1 is noted as absolute insulin insufficiency which

characteristically manifests in younger individuals, brought on by auto-immune-like degradation

of the insulin producing beta-cells located within the pancreas. DMT-2 is of slower onset,

manifesting in older individuals, caused predominately by individual lifestyles. DMT-1 is

managed by the frequent subcutaneous administration of exogenous insulin, possibly in

conjunction with oral medication. In contrast, due to its origin, DMT-2 is mainly managed by

lifestyle changes. Followed by therapies which include oral anti-diabetic medication, as first

choice regime. If these measures are inefficient at addressing DMT-2 pathophysiology

subcutaneous insulin can be employed as add-on therapy (Delamater, 2006:71).

Solid oral dosage forms (SODFs) are preferred, not only in anti-diabetic therapy but also in

other treatment protocols. These dosage forms are easier to administer to conscious patients;

requires little to no organoleptic consideration; needs little aseptic handling; can easily be stored

and transported; and added increased patient compliance with a decreased dosing interval. In

contrast to all these advantages, several disadvantages are also present, which include

administration difficulties for younger children, comatose and unconscious patients, delayed

action before gastro-intestinal absorption, limited dose capacity per dosage, and limited physical

size range of the dosage form. For treatment of diseases, such as diabetes that requires

regular control and monitoring, it is prudent to design a user friendly dosage form with ideally,

no incompatibilities with the patient’s physiological, pathological and lifestyle needs. Due to a

lack of an idyllic setting, an ideal product is not possible; however, researchers have attempted

to design a near perfect product that might fulfill patient related expectations and requirements.

These include, but are not limited to the lower dose load, controlled release of the drug,

affordability, sustainability and versatility (Bardonnet et al., 2006:2).

Current oral anti-diabetic therapeutic regimens include biguanides, e.g. metformin;

sulphonylureas, e.g. gliclazide; and thiazolidinediones, e.g. rosiglitazone. Biguanides are

preferred as a first-line regimen in DMT-2, whereas sulphonylureas are the second class of

treatment in later stage diabetic patients. Due to patient lifestyles and psychologies, patients

rarely pick-up on early symptoms and dismiss pathologies attributed by other factors in their life

(Lebovitz, 1999:1339). As a result, a large portion of diabetics might seek medical attention at

such a stage that first-line therapy might be insufficient and second-line therapy needs to be

initiated to manage symptoms).

Page 19: Cassava starch as modified release excipient in selected

18

Sulfonylureas, effective in the treatment of DMT-2, have been used as anti-hyperglycaemic

therapy since the mid-1950s. For many years this class of active compounds has been one of

the pillars of oral anti-diabetic therapy. Gliclazide is a second-generation sulfonylurea, which

stimulates insulin secretion by closing ATP-sensitive potassium channels in pancreatic beta

cells. It is classified as a weak acidic compound that comprises a larger hydrophobic character

than first generation sulfonylureas. Gliclazide also shows a lower tendency to induce

hypoglycemic episodes in patients. According to Remko (2009:77) gliclazide’s hydrophobicity

makes its activity more effective over an extended time duration. However, it is well known that

insufficient solubility of active compounds may lead to reduced absorption (Dressman et al.,

1998:12). Remko (2009:77) stated that although the second generation sulfonylurea derivatives

(including gliclazide) wereslightly soluble (water solubility of 138.4 mg.l-1 for gliclazide at 25⁰C),

they did depict a fast absorption rate. Through formulating gliclazide into a controlled release

dosage form (once daily dose), it should be possible to extend its activity from a half-life of

approximately 11 h. Characteristically this would increase patient compliance due to fewer dose

intervals (Bartels et al., 2004:9; Remko et al. 2009:77; Vanderpoel et al., 2004:2073).

Starches, which were used in this study are characterised by bio-polymers that have multiple

applications such as fillers, binders and disintegrants in the pharmaceutical and

biopharmaceutical fields. Possible reasons for the use of starches are:

Cost-effectiveness of the starch,

The fact that they are renewable materials,

Available in large quantities,

Non-toxic,

Biocompatible, and

Biodegradable.

In the 1980s it was discovered that certain starches retain unique features that suggest their use

as an excipient for the manufacturing of controlled release SODFs. Due to their versatile

properties, it is possible to obtain quasi-zero-order pharmacokinetic profiles with a very simple

and cost-effective manufacturing process. Tablets produced from starches show low sensitivity

in their release profiles towards manufacturing conditions such as tableting pressure (Chitedze

et al., 2012:32; Lemieux et al., 2009:172; Lenaerts et al., 1991:43). Furthermore, high amylose

cross-linked starch matrix formulations can be manufactured by using conventional tableting

techniques. This kind of technology ranks among the most cost-effective means of

Page 20: Cassava starch as modified release excipient in selected

19

manufacturing controlled release dosage forms for orally administered active compounds

(Lenaerts et al., 1998:229).

Cassava is produced in Latin America, Southern Africa, China, United Arab Emirates and India.

Its standing as a source of starch is rapidly mounting, particularly due to its low price on the

world market when compared to starches from other sources. The potential use of cassava

starch as binder as well as a matrix for the development of edible films has previously been

considered (Chitedze et al., 2012:32; Famá et al., 2006:8; Famá et al., 2007:266). However,

little has been studied on its ability to act as a controlled release excipient in orally administered

formulations (Casas et al., 2010:72).

1.3 OBJECTIVES

In order to achieve the aims of this study, the following will be done:

1) Characterisation of cassava starch with regards to physical properties, powder flow

properties, particle size and morphology.

2) Formulation of beads containing cassava starch as excipient in varying concentrations

(gliclazide, a weak acidic active ingredient (pKa of 5.6) which is poorly water soluble, will

also be included as a model drug).

3) Evaluation of different bead formulations in terms of their physical properties and drug

release profiles.

4) Comparison of different bead formulations, in terms of drug release behaviour, to a

commercially available equivalent (Diamicron®, a readily available gliclazide solid oral

dosage form was selected for this study).

Page 21: Cassava starch as modified release excipient in selected

20

CHAPTER 2

LITERATURE STUDY

2.1 INTRODUCTION

According to the 2013 fact sheet published by the World Health Organisation (WHO), diabetes

is prevalent in approximately 347 million individuals worldwide. Furthermore, the WHO also

estimates that diabetes will be the 7th leading cause of death by 2030 (WHO, 2013). In the

United States alone, an estimated 17.5 million patients were living with diabetes in 2007 at an

estimated cost of US$218 billion (Dall et al., 2010:297). By the year 2000, the health cost

concerning diabetes in sub-Saharan Africa was an estimated US$67.03 billion, both directly and

indirectly to patients and economies within this region. In 2010 an estimated 12.1 million

patients were living with diabetes and an estimate of 23.9 million will be living with diabetes by

2030 in this region alone (Hall et al., 2011:1-2).

Diabetes mellitus is defined as a chronic disease characterised by insufficient glycaemic control,

either due to insufficient insulin production, as in the case of DMT-1, or

tissue-insensitivity and insufficient response to insulin, as in the case of DMT-2. For the

purpose of this study only DMT-2 will be highlighted (Lebovitz, 1999:1339-1340;

WHO, 2013).

In contrast to DMT-1, patients with DMT-2 are of an older demographic and have a slower rate

of onset. The leading cause for DMT-2 is lifestyle dependent factors e.g., insufficient

cardio-vascular exercise, obesity, stress and inappropriate diets. It should also be noted that

genetic and environmental factors contribute to the onset of DMT-2 (Lazar, 2005:374; Lebovitz,

1999:1339-1340).

DMT-2 is described as a dysregulation in insulin and glucose control due to cellular decay of

pancreatic beta-cells; this being a result of over stimulation of these particular cells.

Consequently, these cells are depleted, or completely desensitised to changes in blood glucose

levels. In regards to treatment, all depending on the stage of development, early use of oral

antidiabetic medications can be used to improve glycaemic control (Fowler, 2007:131).

Page 22: Cassava starch as modified release excipient in selected

21

These drugs include:

biguanides, e.g., metformin;

sulphonylureas, e.g., gliclazide, glibenclamide, glipizide;

meglitinides, e.g., repaglinide, mitiglinide;

d-phenylalanine derivatives, e.g., nateglinide;

thiazolidinediones, e.g., pioglitazone, rosiglitazone;

α-glucosidase inhibitors, e.g., acarbose, miglitol;

amylin analogues, e.g., pramlintide;

glucagon-like-polypeptide 1 (GLP-1) analogues, e.g., exenatide, liraglitide; and

dipeptidyl peptidase-4 inhibitors, e.g., sitagliptin, saxagliptin, vildagliptin.

Each of these oral drugs targets either the improvement of insulin secretion, or the improvement

of tissue-sensitivity to insulin. In advanced cases patients might require exogenous insulin

administration in conjunction with oral antidiabetic therapy (Katzung, 2009:737; Lebovitz,

1999:1339-1340; WHO, 2013).

The number of patients diagnosed with chronic and lifestyle diseases such as diabetes, has

increased drastically since the industrial revolution (Cordain et al., 2005:341-344). With the

industrial revolution came a more consumer focused economy. The mechanisation of several

industries, for example agriculture, has led to a less labour intensive economy. This paradigm

shift is dominant in developed economies, e.g. Europe, Japan and North America. Increased

consumerism and decreased physical exertion have led to a more obese population

(WPRO, 2007:1-27).

Obesity is a condition characterised by a higher than normal body mass index and an increase

in plasma lipids. The increase in lipids within tissues influences the metabolic nature of insulin,

credited to the mass number of lipids that needs to undergo lipolysis. This places a strain on

insulin production by the pancreatic beta-cells (Day & Bailey, 2011:55-57).

Patients in developing nations such as sub-Saharan Africa, South America and some South

Asian countries, lack basic health care, education and nutrition. These disparities are present

due to socio-economic, geopolitical and industrial factors (Duraiappah, 1998:2167-2176).

Education, healthcare and nutrition are respectively perceived as basic human rights. Due to

the disparity present in these nations, individuals are deprived of these basic rights (Kawachi

et al., 1997:1491-1498; Wagstaff, 2002:97-102). Insufficient nutrition, may lead to insulin

Page 23: Cassava starch as modified release excipient in selected

22

dysregulation. Instead of maintaining a normal metabolism of glucose and lipids, insulin

production begins to reduce and metabolise protein in the body as a source of energy. This

dysregulation of insulin homeostasis leads to malnutrition associated pancreatic beta-cell

degradation (Taksande et al., 2008:19).

On the other hand, insufficient healthcare or the lack thereof, leads to delayed or wrong

diagnosis. The patient does not receive primary care or education in regards to proper nutrition

and healthcare, for example the identification of symptoms associated with diabetes

(Motala & Ramaiya, 2010:9-36). Due to the disparities in primary health care, these patients’

insufficient diagnosis or delayed diagnosis, palliative care would be considered redundant and

costly. Patients, who do receive any type of treatment, are provided treatment at an

unsustainable cost. SODFs are considered less expensive than any other dosage form, but

even this can amount to unsustainable expenditure on healthcare (Jewesson, 1996:1; Lajoinie

et al., 2014:1088-1089).

2.1.1 TREATMENT OF TYPE 2-DIABETES

The current first line regimen for DMT-2 is oral antidiabetics. This includes biguanides

(metformin), whereas sulphonylureas are considered an add-on, or second line therapy in more

progressive patients (Lahiri, 2012:73; Mcculloch, 2014:1-2). In this study, formulation

strategies, mainly using sulphonylureas, will be the focus.

Sulphonylureas, a second line treatment for early and progressive DMT-2, was first discovered

in the 1950s, with tolbutamide, chlorpropamide, acetohexamide and tolazamide being the model

drugs. Current treatment available is mainly second generation sulphonylureas, for example

gliclazide, glibenclamide and glipizide (Rendell, 2004:1339).

Sulphonylureas’ mechanism of action is based on the closure of the adenosine-triphosphate

(ATP)-mediated potassium ion channels involved in the secretion of insulin by the beta-cells.

Closure of these channels lead to the exocytosis of insulin in response to an increased

concentration of blood plasma glucose. These channels are not completely closed which

prevents possible sulphonylurea induced inhibition at high plasma concentrations (Panten et al.,

1996:1; Rendell, 2004:1339).

Page 24: Cassava starch as modified release excipient in selected

23

2.1.1.1 Gliclazide

Gliclazide (figure 2.1) is poorly water soluble and is rapidly absorbed after oral administration. It

is an intermediate acting hyperglycaemic drug, has a plasma protein binding of approximately

96% and is predominantly metabolised by the hepatic system, making it readily susceptible to

presystemic metabolism. Peak plasma drug concentrations occur within 3 to 4 h after

administration and the drug has a half-life of approximately 12 h.

Figure 2.1: Chemical structure of gliclazide

Table 2.1 reflects the physicochemical characteristics of gliclazide. Gliclazide has proven to

lead to an increase in insulin secretion in long-term treatment regimens. Due to the efficacious

nature of gliclazide in improving insulin secretion, hypoglycaemia is a dominant side-effect and

can be worsened by several drugs, for example aspirin, sulphonamides and alcohol. Other

adverse effects include cardiac dysregulation, cholestatic jaundice, leucopenia, vomiting,

diarrhoea, thrombocytopenia purpura, weight gain, inhibition of alcohol dehydrogenase

enzymes; and even cutaneous symptoms, such as photosensitivity (Fowler, 2007:132).

Due to the short half-life of gliclazide, it is predominantly available as a twice daily dose

regimen. Multiple dosing intervals increase the complexity of patient compliance. In more

complex dosing regimens the possibility of missed doses become more prevalent. With multiple

dose intervals a patient requires a larger amount of units. The increase in the amount of units

needed, may result in an increase in the cost of therapy per patient (Kardas, 2005:722).

Page 25: Cassava starch as modified release excipient in selected

24

Table 2.1: Physicochemical properties of gliclazide (revised from Drugbank.ca and

ChemicalBook.com)

Characteristics Gliclazide

Chemical Formula C15H21N3O3S

Assay ≥ 98%

Form Powder

Colour White

Melting Point ± 163 - 169°C

Molecular Weight 323.411 g.mol-1

pKa (Basic medium) 1.38

pKa (Acidic medium) 4.07

LogP 2.6

Water solubility 1.9 x 10-01 g.l-1

Metabolism Hepatic,

less than 1% is excreted via the urine

Toxicity LD50 = 3000 mg.kg-1

By extending the rate of release, it is possible to extend the presence of the drug in the

circulation, resulting in less dosing intervals and dose units. Consequently, this leads to a

reduction in missed doses and improved adherence to regimes; which ultimately leads to

improved therapeutic outcomes (Kardas, 2005:722). Modified release SODFs are a possible

approach to counter these disadvantages. The rationale of modified SODFs is based on

prolonging the drug present in the blood plasma. By extending drug-plasma levels, it is possible

to reduce the number of doses a patient requires and thus, improves patient compliance.

Page 26: Cassava starch as modified release excipient in selected

25

2.2 SOLID ORAL DOSAGE FORMS

Solid oral dosage forms (SODFs) are perceived as the most dominant drug delivery system

(Jivraj et al., 2000:58; Perioli et al., 2012:621). Various advantages exist that promote the use

of SODFs. These include the following:

Durability during storage and transport.

Ease in physical handling.

Minimal aseptic handling.

Ease of oral administration (Zhang et al., 2003:372; Zhang et al., 2004:371-390).

In contrast to these advantages, several disadvantages can be identified, which include:

Complexation and agglomeration of the various excipients or substances, and

bio-molecules found within the body for example serum albumin.

Administration difficulties in children, comatose patients, and patients with underlying

pathologies for example tumours or constriction of the oesophagus, which in turn makes

it difficult for patients to ingest (Sastry et al., 2000:138; Schiele et al., 2013:937).

Certainly one of the dominant drawbacks of SODFs is the drug susceptibility to various

metabolic processes, as in the case of presystemic metabolism (Dresser et al., 2000:42-

43; Paine et al., 2006:880-881).

To counter the abovementioned disadvantages and improve patient compliance as well as

convenience, researchers and manufactures have attempted several methods to modify the

release of drugs by altering the mechanism whereby the drug is released. This is accomplished

by chemically changing the drug molecule itself or changing the excipients of the product.

Mechanism based modifications include erosion-, diffusion-, dissolution- and osmosis controlled

release mechanisms (Das et al., 2003:12, Patel et al., 2006:58). However, controlled release

mechanisms have their own drawback. In the case of controlled release, the predominant

drawback is dose-dumping. Dose-dumping is the premature release of a drug from the

controlled-release dosage form. This is contradictory to the base rationale for the development

of controlled-release dosage forms (Krajacic et al., 2003:70).

2.2.1 FORMULATION OF SOLID ORAL DOSAGE FORMS

The formulation of a SODF is an important process in providing an acceptable and usable

pharmaceutical product for patients. Formulation is the process by which different constituents

Page 27: Cassava starch as modified release excipient in selected

26

and processes needed to manufacture a SODF, is determined and optimised. In order to

manufacture a SODF for either conventional or modified release of a drug, a number of factors

need to be considered. These include the excipients and manufacturing method (Allen et al.,

2011:2-6).

2.2.1.1 Excipients used in formulations

SODFs, for example tablets, include several excipients in their formulation. Each type of

excipient is incorporated to impart various characteristics or properties to the formulation.

These excipients include fillers, binders, disintegrants, glidants, anti-adherents, etc. (Alderborn,

2007:449; Allen et al., 2011:225). Table 2.2 provides various examples of the different excipient

types which can be utilised for the formulation of SODFs.

Table 2.2: Excipient types and examples

Type of excipient Examples

Fillers

Simple fillers: Microcrystalline cellulose

(Avicel®), micro-fine cellulose, lactose, calcium

phosphate, sugar, dextrose, etc.

Compound Fillers: Avicel® and colloidal sillica,

Avicel® and lactose, Lactose and maize starch,

Lactose and polyvinylpyrrolidone (Kollidon®),

sugars, etc.

Binders Kollidon® 30, 50, 90, VA-64, etc.

Disintegrants Ac-Di-Sol®, Primojel®, Explotab®, Kollidon® CL,

starches (Sta-RX® 1500), etc.

Glidants Magnesium stearate, colloidal sillica, etc.

Acting as a carrier agent for the drug and other excipients, fillers account for the majority of the

dosage form’s weight and volume. This increase in mass and volume allows for a higher

degree of control in regards to handling the drug. It should be noted that in some formulations

where the amount of drug is large enough, the filler might be redundant (Allen et al., 2011:225).

Page 28: Cassava starch as modified release excipient in selected

27

Fillers should fulfil several requirements before they are eligible to be included in a formulation.

These requirements include:

be chemically inert,

non-hygroscopic,

have biopharmaceutical acceptable properties,

have good technical properties,

possess an acceptable taste, and

be cost effective (Alderborn, 2007:449, Allen et al., 2011:225).

Of all the available fillers, none fulfil all of these requirements simultaneously. Due to the

presence of large amounts of filling agent certain properties which include flow rate,

compressibility and porosity of the filler, might be of concern (Alderborn, 2007:449,

Allen et al., 2011:225).

After mixing of the drug with the chosen filler a binder can be added. Adherence of the

individual molecules is achieved by the binding agent’s inherent mechanism of action. Binders

have various mechanisms by which binding occurs, namely:

Overcoming the electrostatic and intermolecular forces,

liquid based bonding,

mechanical interlocking,

the formation of solid bridges between particles after the evaporation of liquids and

natural occurring adhesive and cohesive forces (Alderborn, 2007:452; Allen et al., 2011:225).

Disintegrants, on the other hand, are added to the powder mix to facilitate drug release from the

SODFs after oral administration. Several mechanisms of action are possible for disintegrants.

They are:

swelling of the particles;

electrostatic repulsive forces between the individual particles;

restoration of the particle shape after compression, and

exothermic reactions.

However, there are currently three main mechanisms of importance. The first mechanism is

based on tablet rupture caused by swelling of the individual particles of the disintegrant powder,

after exposure to moisture. Secondly, disintegration can be facilitated by increasing penetration

Page 29: Cassava starch as modified release excipient in selected

28

of moisture through capillary fissures within the outer layers, eventually resulting in

fragmentation of the tablet. The final mechanism by which tablet disintegration can occur is by

deformation of the powder particles; particles with a natural elasticity may return to its previous

shape (Alderborn, 2007:450-452).

Another excipient that can be included into a formulation is a glidant. Glidants are incorporated

in the powder mix to improve flow properties. A glidant’s mechanism of action is based on

lowering the shearing forces between individual particles or changing the electrostatic

interaction between these particles (Faldu & Zalavadiya, 2012:923-924). Sufficient flow is

necessary in direct compression of certain SODF manufacturing, e.g. tablets and multi-

unit pellet systems. Glidants are recommended, if not required, in direct compression, though it

has proven effective and advantageous in wet granulation as well and even mixtures meant for

extrusion-spheronisation of pharmaceutical pellets (Alderborn, 2007:452; Allen et al., 2011:226).

2.2.2 MANUFACTURING METHODS OF SOLID ORAL DOSAGE FORMS

Different manufacturing methods can be used to form a SODF from the aforementioned

constituents. These methods include wet granulation, dry granulation, direct compression and

even extrusion-spheronised beads. Each of these methods is used in different ways, all

depending on the desired outcome or the characteristics of the excipients and drug.

2.2.2.1 Wet granulation

Wet granulation is considered the most cost effective as well as one of the oldest known SODF

manufacturing methods. A homogenous mixture is wetted with a suitable wetting agent (e.g.

water). The moist mixture can be milled or granulated by a granulator in order to form granules.

Prior to tableting, the granules are sieved to homogenise the granule size and to break

agglomerates. The homogenous granules are compressed into a tablet or placed in a capsule.

Wet granulation has several advantages; these include the usability of fine powders, flexibility in

the amount of wetting agents used, and the mixing of powders which do not adhere to each

other. On the other hand, some of the disadvantages include weak cohesion if the wetting

agent dries and did not supply sufficient cohesion between powder particles; and possible

hydrolysis of the excipients or drug (Summers & Aulton, 2007: 412; Tousey, 2002:8-13).

Page 30: Cassava starch as modified release excipient in selected

29

2.2.2.2 Dry granulation

Dry granulation can be used when excipients are for example moisture sensitive. Again, a

homogenous powder mixture is prepared. Two distinct methods can be used to manufacture

SODFs from this method:

Heavy-duty compression of the mixture into a large tablet, and

Roller compression of the mixture between cylinders.

The resulting product is milled to break it into granules. These granules are sieved to form a

homogenous granule size range. Subsequently, the granules are either compressed, or

encapsulated. One of the most prominent advantages of dry granulation is the use of this

method in manufacturing tablets containing moisture sensitive drugs and/or excipients. In turn,

dry granulation is not suitable for fine or physically incompatible powders. Mechanically, this

method of manufacturing has a high level of machine noise (Summers & Aulton, 2007:412;

Tousey, 2002:8-13).

2.2.2.3 Direct compression

A modern method of SODF manufacturing is direct compression of excipient powders into a

single unit. A homogenous mixture of dry powders is introduced into a suitable die via a hopper.

Compression of the mixture occurs by applying force to the mixture present in the die. This

compression is achieved by an automated press and punch. Compression of this powder

causes deformity of the powder particles. In some cases, when the applied force is removed

and the tablet exits the die, the individual particles of the powder might return to its original

shape due to the elastic nature of some of the excipient particles, possibly resulting in capping

or lamination of the solid tablet. These defects decrease the strength and durability of the

tablet. Binders can influence the elastic nature of the powder particles and thus prevent

capping or lamination; maintaining the integrity of the tablet (Alderborn, 2007: 467-473; Tousey,

2002:8-13).

2.2.2.4 Extrusion-Spheronised pharmaceutical pellets

Extrusion-spheronised pellets (beads) are a modern type of SODFs. Beads are manufactured

by extruding a wetted mass of excipients through a perforated screen to form uniform sized

extrusion. These extrusions are then spheronised to uniformly sized and shaped beads by the

use of a multi-bowl spheroniser. These beads can be delivered individually, collectively or

incorporated in a larger unit, e.g. multi-unit pellet tablets or capsules. These individual units are

Page 31: Cassava starch as modified release excipient in selected

30

collectively referred to as multi-unit particulate systems. Multi-unit pellet systems have proven

useful and advantageous in modified release SODFs (Gandhi et al., 1999:161).

2.3 IMMEDIATE RELEASE COMPARED TO

MODIFIED RELEASE SOLID ORAL DOSAGE

FORMS

The basic rationale of any drug release from a SODF is to provide an adequate plasma

concentration of the drug. This level falls in a concentration range; ranging from the minimum

therapeutic concentration to the minimum toxic concentration and this range is known as the

therapeutic index. Any drug concentration below the therapeutic index is sub-therapeutic,

whereas any concentration above the index is toxic. Immediate release of a drug is identified by

an initial release of drug which peaks after a certain time (relatively short) has passed. After the

peak is reached, the concentration level drops. In order to maintain a suitable therapeutic

concentration of the drug, the next dose needs to be timed correctly in accordance with the drug

half-life. A disadvantage of note is that with immediate release, sub-therapeutic or a toxic level

of the drug is possible. To counter this, modified release dosage forms are continuously being

developed. The rationale of modified, sustained or controlled release dosage forms is to

provide a constant plasma drug concentration over a prolonged time period. This extension of

drug present in the blood plasma reduces the number of doses required to provide a therapeutic

drug concentration (Allen et al., 2011:258). This described rationale for immediate drug release

compared to modified drug release is illustrated in figure 2.2.

Figure 2.2: Graph comparing immediate and controlled modified drug release

Page 32: Cassava starch as modified release excipient in selected

31

2.3.1 TYPES OF IMMEDIATE RELEASE SOLID ORAL DOSAGE FORMS

2.3.1.1 Conventional release solid oral dosage forms

The release of a drug from a conventional release SODF is characterised by the

physicochemical properties of the drug and dosage form. Conventional release SODFs are

known to release a drug as it transits through the body. These dosage forms are basically

administered as a unit-dose, implying for example that one tablet contains a specified dose

amount of the active agent. The tablet is easily administered by means of oral intake. Once the

tablet enters the gastrointestinal tract, it is exposed to gastrointestinal fluid, enzymes and other

biological factors. Transit is necessary for the disintegration and dissolution of the tablet, which

releases the drug from the SODF. The fluid present in the body saturates the tablet and

saturation allows for the incorporated excipients to fracture the solid tablet into smaller pieces.

A decrease in particle size leads to an increase in the surface area exposed to the fluid

environment; this increase allows for an increased rate of disintegration. After disintegration of

the SODF to a smaller particle size range; the particles will undergo dissolution. Once in

solution, the active agent can cross the epithelium into the blood stream, depending on the

permeability of the drug. At this stage the drug travels along the circulatory system to the site of

action. Both disintegration and dissolution can be described as a rate limiting step in the

intended release of a drug. The rate of these different steps can be influenced by the various

manufacturing methods, the choice of excipients, or the formulation itself. Other factors can

influence these steps as well, such as biological, environmental and physicochemical factors,

for example particle size of the excipients and water solubility. Reasons in favour of

disintegrating tablets include ease of administration, predetermined dose size, and patient

compliance. However, several drawbacks prevent the use of this type of tablet, which include

comatose patients, pathologies of the ora-esophogeal tract, and young children or elderly

individuals with difficulties in swallowing (Allen et al., 2011:225-226; Sahoo, 2007:20-31).

2.3.1.2 Effervescent Tablets

Effervescent tablets are designed to include a higher amount of drug; and most importantly,

disintegrate and dissolve within a glass of water. Key ingredients in the design of effervescent

tablets are bicarbonates or carbonates, and citric and/or tartaric acid, which in combination form

part of the disintegration system. As the effervescent tablet is exposed to water, it starts to

permeate the tablet. This in turn causes a reaction between the carbonate and acid which

produces carbon dioxide. Release of carbon dioxide disintegrates and dissolves the tablet. The

Page 33: Cassava starch as modified release excipient in selected

32

solution that is formed allows for a rapid onset of action and rapid emptying of the

gastrointestinal tract. By dissolving the SODF into a solution, it is possible for patients with

underlying pathologies which limit the intake of other SODFs, to ingest the medication. Elderly

and young children are also benefitted by using this type of SODF. One of the limitations of this

delivery system, however, is the organoleptic properties of the SODF excipients present in

solution, particularly the flavour of the solution. Due to the large amount of ingestible liquid, it is

recommended that the dosage form contains a flavouring agent (Alderborn, 2002:412;

Alderborn, 2007:456; Allen et al., 2011:228).

2.3.1.3 Chewable Tablets

Some SODFs are mechanically crushed by means of chewing. This ensures the complete

disintegration of the SODF into smaller particles. Though it should be noted that dissolution

does not fully occur in the mouth, but still in the gastrointestinal tract, this acts as the

disintegration process needed for drug delivery within the gastro-intestinal tract. As a result, this

allows for a faster dissolution of the SODF, and thus faster absorption. Due to the prolonged

presence within the mouth, flavouring is yet again a concern. If the patients do not prefer the

flavour of the tablet after chewing, it will affect patient compliance negatively (Alderborn,

2002:412; Alderborn, 2007:456; Ansel et al., 2011:227; Siewert et al., 2003:3).

2.3.1.4 Sublingual and Buccal Tablets

Another SODF that releases the drug immediately is sublingual and/or buccal tablets.

Sublingual tablets are SODFs which dissolve under the tongue, whereas buccal tablets dissolve

on the inside of the cheek or under the lip. The anatomical locations were both sublingual and

buccal tablets function can be seen in figure 2.3. These SODFs are designed to dissolve in the

mouth and be absorbed through the oral mucosa. Once these SODFs dissolve in the mouth, it

should not be swallowed. Again, these dosage forms rely on organoleptic considerations,

especially flavouring. A disadvantage of sublingual and/or buccal tablets is the limited dosage

size, due to the limited absorption capacity of the oral mucosa (Alderborn, 2002:413; Alderborn,

2007:457; Allen et al., 2011:227).

Page 34: Cassava starch as modified release excipient in selected

33

Figure 2.3: Sublingual and buccal route of administration (intranet.tdmu.edu.ua)

2.3.1.5 Multi-layer tablets

The basic concept of conventional multi-layer tablets is based on the repeated compression of

multiple layers containing incompatible active ingredients. It is also an acceptable practice to

colour the various layers, resulting in a uniquely identifiable product (Alderborn, 2002:412;

Alderborn, 2007:456).

2.3.1.6 Lozenges

Lozenges are tablets designed to slowly dissolve in the mouth. They are designed to either

have a local or systemic effect. Once lozenges are in the mouth, the saliva supplies the

necessary fluid which induces dissolution of the tablet and release of the drug. These tablets

can, however, also act as a simple slow release dosage form (Alderborn, 2002:413; Alderborn,

2007:457).

2.3.2 MODIFIED RELEASE SOLID ORAL DOSAGE FORMS

The rational by which modified release SODFs function is based on prolonging the presence of

the active ingredient in the blood plasma. This extended time of the drug present in the blood

plasma improves patient compliance and therapeutic outcomes. This is achieved by lowering

the number of doses required for the patient to maintain a therapeutic drug concentration

(Siegel & Rathbone, 2012:19-20).

2.3.2.1 Coated tablets

An approach to modified release SODFs is the coating of disintegrating tablets. Various

methods of coating were developed for maximum patient convenience, including enteric,

Page 35: Cassava starch as modified release excipient in selected

34

gelatine and film coatings. Each coat is applied using a spaying-dry method. After spraying the

tablet with the coating, the product is dried. One of the main reasons for applying a coating is to

improve dosage forms resistance in low pH environments. This is advantageous in the case of

a drug which is pH sensitive or the location of drug absorption is based in an environment with a

high pH (e.g. the intestinal tract) (Alderborn, 2002:412, Alderborn, 2007:456, Ansel et al.,

2011:227, Das et al., 2003:14).

2.3.2.2 Diffusion-controlled tablets

Diffusion-controlled release SODFs rely on moisture permeating it with subsequent drug

release. Diffusion-controlled release dosage forms are divided into two types, namely, matrix

and membrane types. In order for this system to function properly, the dosage form needs to

remain intact while in transit through the gastrointestinal tract. Upon exposure to moisture the

dosage forms starts to release the drug from the matrix or membrane which encompasses the

drug. Depending on excipients and manufacturing process used to manufacture this particular

SODF, the rate of drug release can be augmented to prolong drug release (Uhrich et al.,

1999:3183-3189).

2.3.2.3 Dissolution-controlled tablets

Dissolution-controlled release relies on the dissolution of poorly water soluble salts of the active

agent, using a slowly dissolvable carrier or covering of the drug particles with a slowly dissolving

coating (Uhrich et al., 1999:3183-3189).

2.3.2.4 Erosion controlled tablets

These tablets are a single unit system consisting of a matrix based structure. The active agent

is dispersed throughout the matrix. As the matrix starts to dissolve, the active agent is released.

This erosion leads to a loss in tablet weight and a predictable release profile of the active agent

(Colombo et al., 2000:201-202; Dey et al., 2008:1069).

2.3.2.6 Osmosis controlled tablets

Osmosis controlled release is based on a difference in osmotic pressure between the interior

and exterior environment of the dosage form. A semi-permeable membrane is permeated by

moisture due to this osmotic pressure difference. The active ingredient within the dosage form

starts to dissolve and the resulting solution is then pumped out of the dosage form via a single

orifice or through a semi-permeable membrane. This transport is a convective transport

Page 36: Cassava starch as modified release excipient in selected

35

process. Several pump mechanisms can be utilised, which include the introduction of a swelling

layer that forces the solution out as the layer expands. Another method is that the solution itself

exhibits swelling properties. Each of these methods produces pressure, thus forcing the

solution through the orifice. Osmosis controlled systems can be manufactured as a single- or

multi-dose system (Dey et al, 2008:1069; Gupta et al, 2010:571-582). Figure 2.4 illustrates an

example of an osmotically controlled release tablet.

Figure 2.4: Example of an osmotically controlled release tablet

2.3.2.7 Multi-layer tablets

Multi-layer tablets contain layers composed of different drug concentrations per layer; or each

layer is compressed to various degrees of density and strength. In the case of varying drug

concentration, upon the disintegration and dissolution of each layer, a different amount of the

active ingredient is released at various stages during gastrointestinal transit. If the

concentration of the drug is constant throughout the layers, the density of each layer influences

the rate of disintegration and therefore prolongs the release of the drug from the solid dosage

form (Alderborn, 2002:412; Alderborn, 2007:456). These multi-layer tablets are made by

compression of an initial amount of powder mix which is introduced into the die. After

compression the die is filled again with another layer. As a result of the applied force, the first

layer is compressed more densely than with the first compression. This delivers a denser and

mechanically stronger first layer. A slight variation on this method is an initial high pressure

compression of the first layer and then followed by consequent layers where each layer has a

reduced compression force applied to the layer (Abdul & Poddar, 2004:160-161).

Page 37: Cassava starch as modified release excipient in selected

36

2.3.2.8 Multi-particulates

A contemporary approach to modified controlled release dosage forms is the use of

multi-particulate components, which is a system constituted out of smaller individual units with

identical characteristics and properties. Multi-particulates have many advantages which make

them a suitable choice for controlled release, namely:

improved gastric emptying;

easily adjustable dosing;

multi-phase release profiles;

improved flow properties;

decreased dust and powder waste;

decreased tendency for dose dumping to occur;

reduction in both the dose frequency and dose size;

uniform transit through the gastrointestinal tract;

lower tendency to gastrointestinal irritation;

reduced individual variations;

possible multi-drug combinations;

lowered tendency for side-effects;

cost effectiveness;

provide a targeted and controlled release and

a shorter lag time.

This system of a single unit is useful in the case where varying concentrations of a drug need to

be present in a single unit, either a tablet of capsule. Individual particles can be designed with

different concentrations. Another advantage of multi-particulates is that incompatible drugs can

be incorporated into a single unit. The multi-particulates or pellets after manufacturing can now

be directly compressed into a single unit-of-use; or the pellets can be incorporated into a

capsule as in the case of this study. Several methods of multi-particulate manufacturing exist

(Ganhdi et al., 1999:160-161; Khan et al., 2014:2137-2140; Vervaet et al., 1994:131-132; Young

et al., 2002:87-92). These include:

layering,

freeze pelletisation,

cryopelletisation,

hot-melt extrusion and

Page 38: Cassava starch as modified release excipient in selected

37

extrusion-spheronisation.

2.3.2.8.1 Layering

Layering or coating is based on deposition of successive layers of an active ingredient. These

layers are deposited on a core. This core can be a crystal, an inactive agent or granule (Hirjau

et al., 2011:210). The following figure (figure 2.5) provides a diagrammatic representation of the

layering process.

Figure 2.5: Layering process for pharmaceutical beads (revised from slideshare.net)

2.3.2.8.2 Freeze pelletisation

Freeze pelletisation on the other hand is a manufacturing method where spherical matrix pellets

containing the drug are produced. A molten droplet containing the drug and excipients is

introduced into a temperature regulated column containing an immiscible liquid. .The column

consists out of various temperature regions, ranging from -40°C to 100°C. The liquid chosen for

the process needs to have a lighter density then the droplet. This difference in density between

the liquid and droplet allows for a natural conveyance of the droplet through the liquid. As the

droplet “drops” down through the liquid, it moves through the various temperature regions,

consequently forming a solid pellet. Layer by layer the pellet continues to form until it enters a

low temperature region (0°C to -40°C), at which time the deposited layers start to freeze and

solidify. This is an inexpensive and easily reproducible method of manufacturing pellets,

depending on the variables (Cheboyina & O’Haver, 2004:98-102; Lavanya et al., 2011:1345).

Page 39: Cassava starch as modified release excipient in selected

38

2.3.2.8.3 Cryopellitisation

Pellets produced via cryopelletisation, is when a droplet of organic or aqueous liquids are

conveyed through a perforated plate in the presence of liquid nitrogen and a solid pellet is

formed. The shape of the pellets is determined by the distance between the perforated plate

and the nitrogen reservoir. Pellet size is determined by the diameter of the perforations present

in the plate (Gandhi & Baheti, 2013:1624; Lavanya et al., 2011:1344).

2.3.2.8.4 Hot-melt extrusion

Hot-melt extrusion is a solvent free method, ideal for drugs that are unstable in the presence of

moisture. Several processes are used to form pellets by means of hot-melt extrusion and these

processes are:

Plastisation or melting of a drug dispersed throughout a solid medium which acts as a

thermal carrier.

Use of an extruder in order to shape the molten content.

Spheronisation at high temperatures to form uniform spheres.

Solidification of spheres into the desired shape (Lavanya et al., 2011:1345; Patel et al.,

2010:81-82; Young et al., 2002:87-92).

2.3.2.8.5 Extrusion-spheronisation

Extrusion-spheronisation is a multi-phase method of manufacturing, first developed in the

1950s. First, a homogenous powder mixture is wetted with a suitable wetting agent; for

example water. The resulting wet mass is introduced into an extruder. Once introduced into the

extruder, the mass enters a chamber via a hopper. The chamber contains multiple cylinders

which rotate at pre-set rotations and a perforated screen. As the cylinders rotate the mass is

pushed against the screen. Due to the sheering forces and compression of the mass against

the perforated screen, it is extruded through the screen. Figure 2.6 provides an approximate

idea of how the extruder functions. The size of the extrusions is determined by the diameter of

the perforations present in the extrusion screen. Finally, spheronisation of the extruded material

in a spheroniser is conducted. The spheroniser consists of a multi-bowl chamber with an

attached friction plate. As the extrusion enters the bowl, the rotating friction disk and supplied

compressed air create a rotation of the extrusion mass in such a manner that the extrusions

break into smaller sizes and produce spherical pellets. The size and shape of the final pellets

are determined by the rate at which the spheroniser rotates (Newton et al., 1995:101; Vervaet

et al., 1995:136; Young et al., 2002:87-92). For the purpose of this study and the cost-effective

Page 40: Cassava starch as modified release excipient in selected

39

nature of this method it was opted to use extrusion-spheronisation as the method of choice for

the manufacturing of the beads.

Figure 2.6: Radial extruder (revised from spheronizer.com)

2.4 STARCH AS A VERSATILE EXCIPIENT

Starch has proven versatile and invaluable in dosage form design. Due to the flexible nature of

starch and its various applications in the pharmaceutical industry, it is essential to investigate its

application in the design of modified release dosage forms (Dumoulin et al., 1998:161-162;

Ispas-Szabo et al., 1999:163-165; Lenaert et al., 1998:225). Starch, which is a natural

occurring polymer, has a multitude of applications in the pharmaceutical industry; it may be

employed as a filling agent, binder, disintegrant or even as a glidant (Bayor et al., 2013:17). It

has become a necessity to investigate applications of renewable sources for excipients. Starch

is considered a viable candidate in improving the release profile of an active ingredient and

resulting therapeutic outcomes (Dumoulin et al., 1998:161-162; Ispas-Szabo et al., 1999:163-

165; Lenaert et al., 1998:225).

Two principal polymers, amylopectin and amylose present in starch make for an ideal candidate

in the design of controlled release dosage forms. Figure 2.7 shows a structural comparison of

the two distinct polymers. These two polymers form a robust polymer-matrix. An important

property of starch powders is its tendency to gelatinise when moistened. This proves useful in

designing a dissolution- or erosion-controlled release tablet. When the mass is introduced to a

moisture rich environment, it begins to expand as it absorbs the moisture. This occurs as the

branched polymers expand and moisture permeates the polymer-matrix. The mass becomes

gelatinous and forms a pseudo-suspension or matrix. As the starch travels through the

gastrointestinal tract, metabolic processes start to dissolve the mass; this dissolution of the

Page 41: Cassava starch as modified release excipient in selected

40

starch allows for release of drug particles from the polymer matrix. An initial dose of the active

ingredient is released when the matrix starts to dissolve (Mandal et al., 2009:1348). Continuous

dissolution of the mass and release allows for sustained release of the drug (Mandal et al.,

2009:1348). As an abundant source of starch, investigation has been warranted in the possible

application of refined cassava starch as modified release filler.

Figure 2.7: Molecular and macroscopic structure of amylose and amylopectin

(revised from voer.edu.net)

Page 42: Cassava starch as modified release excipient in selected

41

2.4.1 CASSAVA

The arrival of European explorers in the “new world” has meant the sporadic spread of fauna

and flora across the globe. This migration led to many new discoveries within the indigenous or

non-indigenous environments (NOAA, 2008). A good example of this is

Manihot eschulenta Crantz (figure 2.8). Manihot eschulenta Crantz, otherwise known as yuca,

tapioca or cassava, is a perennial root of the Euphorbiacea-family, native to tropical and sub-

subtropical climates as seen on the South-American, South-Asian and the sub-Saharan Africa

continent. Although native to humid climates, this root is quite adaptable to various

environments (FAO, 2013:6-7).

Figure 2.8: Illustration of cassava plant and root (revised form theglyptodon.com)

Cassava is also globally known as one of the main sources for starch with an estimated global

production of 290 x 106 ton in 2012 (FAO, 2013:6). As a source of energy, cassava proves to

be an invaluable staple in the diet of several developing nations. Due to the relative short

lifespan of post-harvested roots, the refined starch powder lengthens the lifespan of the starch.

The process of refinement ensures a thriving economy, not only for subsistent farmers, but also

Page 43: Cassava starch as modified release excipient in selected

42

for refinement centres and post-refinement trading. Refinement is also crucial in improving the

safety profile of cassava starch, due to the presence of cyanide in the cassava root (Fáma et al.,

2006:8; Fáma, et al., 2007:266).

As stated before, cassava starch contains two distinct glucose derived polymers, linear and

helical amylose as well as a short chain branch amylopectin, in ratios of 1:3 – 1:4 (Charles

et al., 2005:2718). Amylose content ranges between 15.0 – 25.0% (Charles et al., 2005:2117;

De Floor et al., 1998:62; Moorthy et al., 2002:560-562; Nuwamanya et al., 2010:1;

Rollande-Sabaté et al., 2012:161). The physicochemical compositions of the aforementioned

polymers form a robust matrix. This natural matrix influences several important aspects of the

starch’s physicochemical properties. The level of crystallinity is directly affected by the number

of hydrogen bonds. If the crystalline structure shows a high level of rigidity, it would indicate a

high number of hydrogen bonds. With a high level of crystallinity, the more robust matrix would

show a tendency to lower fluidity and adversely affect physicochemical properties (Huang et al.,

2007:133). Other properties are highlighted in both tables 2.3 and 2.4.

Tabel 2.3: Content Properties (Revised from Moorthy, 2002:560,561)

Properties Cassava

% Yield from H2O-NH3 extraction medium 21.80 ± 0.540

%Total amylose extracted 0.37 ± 0.010

% Moisture content 10 – 14

% Fibre/Ash Content 0.01 - 0.8

% Lipid content 0.01 - 1.54

% Phosphorus content 0.01 - 0.01

Colour White

Granule shape Round, truncated, cylindrical, oval, spherical,

compound

Page 44: Cassava starch as modified release excipient in selected

43

Granule size 3 - 43 µm

Table 2.4: Physicochemical properties of cassava variants (Revised from Moorthy, 2002:569)

Variant Granule

size

[µm]

Reducing

values

Amylose

content

Past*

temp

[°C]

Vis**

2% paste

Swelling

volume

[ml.g-1]

Sol***

[%]

M-4 5.4 - 35.1 1.8 0.530 60.7 58.0 30.5 22.8

Kalikal

an

5.4 - 40.5 1.8 0.550 63.70 58.0 38.8 24.8

H-1687 5.4 - 40.5 1.4 0.540 55.68 58.0 25.5 23.6

H-2304 5.4 - 43.2 1.4 0.525 52.68 55.0 30.5 24.8

H-226 5.4 - 43.2 1.8 0.500 55.66 56.0 33.8 27.8

H-97 5.4 - 43.2 1.2 0.535 58.70 55.0 30.5 17.2

H-165 8.1 - 48.6 1.6 0.505 52.65 54.0 37.8 27.2

*Pasting, **Viscosity, ***Solubility

Cassava starch has a variety of applications in more than one industry. In the textile industry it

is used in clothing dye. The pharmaceutical industry utilises it as a versatile excipient, for

example fillers. Starch is used in the adhesive, rubber and foam industry. In the paper industry,

cassava starch is also utilised to improve the colour and paper quality of paper stocks. Organic

sugars and acids can be derived from cassava starch. Fructose syrup and gelatine capsules

can also be produced using sugars prepared from cassava starch. Employing bioreactor

processes which incorporate Aspergillus awamori and Lactococcus lactalis spp. lactis, L-lactic

acid can be produced. Phytase production is also possible with cassava starch (Fao, 2005;

Tonukari, 2004:5-6).

As observed in table 2.4, swelling is an important characteristic of cassava starch. The

tendency of the polymers to swell when in contact with moisture is of noteworthy importance in

the possible manufacturing of modified SODFs. Being susceptible to digestive processes, the

mass is dissolved in the gastrointestinal tract (Beneke et al., 2009:2612-2614). Dissolution of

the mass releases the drug from the resulting gelatinous mass. Due to the availability and

Page 45: Cassava starch as modified release excipient in selected

44

inexpensive nature of cassava starch, it has been deemed a promising candidate in the pursuit

of a cost effective and a renewable excipient in the production of modified release SODFs

(Dumoulin et al., 1998:361-362; Lenaerts et al., 1998:233-234). Though the versatility and its

renewability promises cassava starch to be a viable candidate for pharmaceutical product

manufacturing, scrutiny regarding the patient safety e.g. allergies and toxicity, would need to be

investigated. For the purpose of this study, this line of enquiry was forgone.

2.5 SUMMARY

In this chapter diabetes was briefly discussed, as well as one of the most dominant second line

oral antidiabetic drugs, gliclazide. An overview of different factors necessary in the formulation

of either immediate or modified release SODFs was also provided. Furthermore, starch as a

versatile and matrix-rich excipient in SODFs, specifically cassava starch as an inexpensive and

renewable source of starch, was discussed. It is this rationale that warrants the evaluation of

cassava starch as a suitable excipient in modified SODFs. In chapter 3, the materials and

methodology employed for this study will be discussed.

Page 46: Cassava starch as modified release excipient in selected

45

CHAPTER 3

EXPERIMENTAL METHODS AND MATERIALS

3.1 INTRODUCTION

Any pharmaceutical dosage form, conventional or specialised, is formulated in order for a

patient to receive an effective drug dose. Appropriate design and formulation require

methodical understanding of the functional factors that affect the physicochemical

characteristics of the drug and excipients used, as well as the absorption of the drug. The drug

and excipients incorporated into the formulation have to be compatible in order to produce a

product that is stable, efficient, striking, easy to administer, and safe. Furthermore, formulation

of a solid oral dosage form usually necessitates accurate processing control of the powder

mixture to guarantee a homogeneously formed product. Numerous excipients are

gravimetrically added to form the bulk powder with which homogeneity is accomplished through

optimum mixing. Homogeneity during tablet manufacturing is also accomplished through the

correct process used to achieve good flow of the mixture into the tablet die. Extrusion-

spheronisation was used in this study to increase the bulk density and increase flowability of the

formulations (Aulton & Taylor, 2013:480; Shah & Mlodozeniec, 1977:1377). Thus, in order to

evaluate which formulation is most appropriate, it is of utmost importance to evaluate the above-

mentioned factors influencing design and formulation.

This chapter deals with the pharmaceutical excipients (materials) used in the various

formulations tested. Moreover, it describes the experimental procedures employed to determine

the effect of these excipients on the physical properties of the beads formulated as well as on

the dissolution profiles of the formulations.

3.2 MATERIALS

The pharmaceutical materials employed in this study, their respective batch numbers as well as

where these materials were sourced, are presented in table 3.1. All of the materials were of

analytical grade and were used as supplied.

Page 47: Cassava starch as modified release excipient in selected

46

Table 3.1: Pharmaceutical materials employed in the various formulations, batch numbers and suppliers

Materials Batch nr. Source

Gliclazide 100111302045 Bal Pharma, Ltd. Bengaluru, India

Cassava starch 169A-27-11-12 Meelunie, BV.

Amsterdam, Netherlands

Cassava starch

(Mbundumali-namwera) Donated Malawi

Consolidated Starch 23871 Warren Chem Specialities Cape Town, South Africa

Avicel® pH 200 M939C FMC International,

Wallingstown, Ireland

Kollidon® 30 8608522440 BASF, SE.

Ludwigshafen, Germany

Hydroxypromethylcellulose 11040 Shin-Etsu Chemical, Ltd.

Tokyo, Japan

Hydrochloric acid 44836 Saarchem, Ltd.

Krugersdorp, South Africa

Methanol L361202 VWR International, Ltd.

Poole, England

Ethanol 180914ET Rochelle Chemicals, Cc.

Johannesburg, South Africa

3.3 CHARACTERISATION OF CASSAVA STARCHES

The physical properties of powders have a significant effect on the flowability and tabletability of

formulations. The primary physical excipient properties of importance are moisture content,

particle size and particle size distribution. Other properties (which are derived from the primary

properties) include flowability, compactibility and compatibility. The properties that were

evaluated during this study are described in the following sections.

Page 48: Cassava starch as modified release excipient in selected

47

3.3.1 THERMOANALYTICAL CHARACTERISATION

The presence and distribution of moisture depend considerably on the chemical nature of a

certain material, its physical properties such as particle size and porosity; and on the ambient

relative humidity (RH), which determines the equilibrium moisture content (Garr & Rubinstein,

1992:187-192; Teunou et al., 1999:109-110). Moisture may have noteworthy effects on the

density of materials, flowability, binding characteristics, lubrication properties, compression,

surface tension, tablet tensile strength and tablet toughness (Teunou et al., 1999:109-110;

Viljoen et al., 2014:731-741).

Thermoanalytical characterisation of the cassava starch powders was conducted at various time

intervals (30; 60; 120; 240; 360 and 480 min) and at various temperatures

(25; 30; 40; and 50°C).

3.3.1.1 Differential scanning calorimetric (DSC) analysis

DSC is used to determine physical properties based on thermal transition. Thermal stability of a

substance at increasing temperatures and specific time intervals can be evaluated using

DSC-analysis (Roy et al., 2002:399-400).

A Shimadzu DSC-60A (Shimadzu Scientific Instruments, Shimadzu, Japan) instrument was

used to obtain DSC-spectra of the cassava starch samples. Approximately 2 mg of each

sample was weighed into aluminium pans. These pans were sealed with a lid; each lid was

crimped in place by using a Du Pont crimper. The lids where pierced to form a small pinhole in

order to elevate possible pressure build-up within the pans. A similar sealed, empty pan was

used as reference. DSC-spectra were obtained at a heating rate of 10°C.min-1 under a nitrogen

purge of 30 cm3.min-1. The individual spectra were determined up to a temperature of 300°C

(Lemmer et al., 2012:331; Viljoen et al., 2014:732).

3.3.1.2 Thermogravimetric analysis

Thermogravimetric analysis (TGA) is based on the change in weight of a sample at various

temperatures (Ko et al., 2014:155).

TGA was conducted on each starch sample at a temperature range of 0 - 300°C.

TGA thermograms were recorded with a Shimadzu DTG-60 instrument (Shimadzu, Kyoto,

Japan). The weight of each sample was approximately 5 - 8 mg and heating rates of 10°C.min-1

under nitrogen gas flow of 35 cm3.min-1 were used. The theoretical weight loss during the

Page 49: Cassava starch as modified release excipient in selected

48

different conditions for each sample was calculated (in percentage) and compared.

Equation 3.1 applies to stoichiometric reactions with just partial weight loss such as dehydration.

100%mMn

MΔG

0Gas

m

[3.1]

Where the percentage content (G) is calculated from the weight loss (Δm) and the initial sample

weight (m0). M is the molar mass of the sample tested; MGas is the molar mass of the gas

liberated and n is the number of molecules liberated per starting molecule (Lemmer et al.,

2012:331; Viljoen et al., 2014:732).

3.3.1.3 Karl-Fischer titration

Moisture content for each sample was determined with a Mettler DL 18 Karl-Fischer titrator

(Mettler Toledo International LLC, USA). The Karl-Fischer solution was calibrated against a

predetermined mass of water. An accurately weighed (250 mg) cassava starch sample was

added to ethanol which was neutralised with the Karl-Fischer solution beforehand in a titration

beaker. The mixture was magnetically stirred and titrated with the Karl-Fischer solution.

Experiments were conducted in duplicate and the percentage water (w/w) calculated as follow:

10050W1000

CBMMoisture %

[3.2]

Where M is the Karl-Fischer titrant volume (ml); B is the volume (ml) of Karl-Fischer titrant for

the blank; C is the calibration amount (mg H2O.ml-1 Karl-Fischer titrant) and W is the sample

weight in grams (Aucamp et al., 2013:20; Viljoen et al., 2014:732).

3.3.2 INFRARED (IR) ANALYSIS

Vibrational characteristics, e.g. stretching and bending of different molecular bindings can be

examined and identified with the use of infrared (IR) spectrometry. Compounds can even be

identified using these IR-spectra due to individual compounds having distinctive IR-spectra

(Chistian, 2004:469-472; de Kock, 2005:60).

IR-spectra of the cassava starches were recorded on a Nicolet Nexus 470 FT IR ESP

spectrometer (Thermo Fischer Scientific, Waltham, Massachusetts, USA) over a range of

4000 - 400 cm-1 using the potassium bromide (KBr) referencing technique. Small samples of

approximately 2 mg were collected at different temperatures and time intervals; and individually

Page 50: Cassava starch as modified release excipient in selected

49

mixed with 200 mg KBr (Merck, Darmstadt, Germany) prior to analyses. This analysis was

repeated (with identical parameters, excluding the KBr reference) with a Bruker® Alpha Platinum

FT-IR Spectrometer (Bruker®, Billerica, Massachusetts, USA) in order to provide spectra with a

higher resolution (Aucamp et al., 2013:20; Lemmer et al., 2012:331).

3.4 SOLID ORAL DOSAGE FORMS

SODFs are presently the most preferred dosage form globally. The preference for SODFs is

accredited to the many advantages, for example: improved patient compliance due to ease of

administration, ease of transport, long shelf life and cost effective manufacturing (Alderborn,

2007:455-456; Hirani et al., 2009:162; York, 2013:7-8). As described in chapter 2, SODFs

consist of various excipients and the active pharmaceutical ingredient (drug). Each of these

ingredients is included in a formula for the manufacturing of a specific SODF. These ingredients

comprise, but are not limited to; fillers, binders, glidants and disintegrants, and most importantly

the drug. The drug and the different excipients as well as varying amounts of each of these

constituents need to be incorporated into a basic formula in order to be able to manufacture a

SODF.

3.4.1 PREPARATION OF BEADS

Pharmaceutical pellets (beads) are a modern method of SODF manufacturing and have proven

useful in the application for modified release dosage forms as well as the improvement of drug

release and physicochemical characteristics for example flow or physicochemical compatibility

between different drugs (Gandhi et al., 1999:160-162, Vervaet et al., 1994:131). The table

(table 3.2) below represents the factors and levels required to design the necessary

experimental formulations.

Table 3.2: Variables and different levels of each variable as employed in this study.

Factor

Levels

0 1 2

Drug (Gliclazide) 5% (w/w) 10% (w/w) 15% (w/w)

Filler Avicel® PH 101 Cassava starch Not applicable

Binder: Kollidon® 30

0% (w/w) 3% (w/w) 5% (w/w)

Page 51: Cassava starch as modified release excipient in selected

50

Polymer: HPMC

0% (w/w) 5% (w/w) 10% (w/w)

Powder mixtures were prepared. The composition of these mixtures were determined using a

partial factorial design. The composition of these mixtures were noted in table 4.2. The filler,

drug and binder for each formulation were weighed (mixture weighing 100 g) and transferred to

a glass bottle. Each of these bottles was covered with Parafilm® before closure with a screw

cap. The powders were mixed using a Turbula®-mixer (Model T2C, W.A., Switzerland) at

69 rpm for 10 min. After mixing, each powder mixture was wetted using a 70:30 deionised

water and ethanol mixture. The wetted mass was mixed using a mortar and pestle. After each

5 ml volume of the wetting agent added, the mixture was blended with a blender (Model FP731

Multi-Pro, Kenwood®, South Africa) for approximately 2 min. This was repeated till the correct

consistency was acquired. Upon completion of the addition of the wetting agent, the wetted

mass was passed through an extruder (Caleva® Extruder 20, Sturnminster Newton, England),

with the roller speed set at 32 rpm. A 1 mm diameter perforated screen was employed during

the extrusion. The resulting extrudate was spheronised in a multi-bowl spheroniser at 3000 rpm

for 10 min (Caleva®, Sturnminster Newton, England) (Chinyemba, 2012:21; Mallepeddi et al.,

2010:54). After spheronisation, beads were formed and the beads were dried at 40°C for 24 hr.

Bead samples were sieved to provide a mono-dispersed size range.

3.4.1 MORPHOLOGY OF POWDER PARTICLES AND BEAD

FORMULATIONS

Morphology is defined as: “the study of the forms of things, in particular” (Oxford dictionary) and

with the investigation of powder, bead and tablet morphology, various macroscopic phenomena

can be observed (de Kock, 2005:62). Differences between powder formulations, especially in

terms of flowability, packing formation and compression can be explained through differences in

their morphology. It is therefore important to investigate particle shape and size in order to be

able to predict for example, powder flow and packing arrangement (Hancock et al., 2004:980;

Lavoie et al., 2002:892; Velasco et al., 1995:2385).

3.4.1.1 Scanning electron microscopy (SEM)

Scanning electron microscopy (SEM) was used to identify the particle shape and surface

structure of the different starches used and bead samples that were prepared in this study.

Page 52: Cassava starch as modified release excipient in selected

51

SEM analysis provides information on microscopic level to better understand the macroscopic

behaviour of a powder or bead formulation (de Kock, 2005:62).

Each starch and bead sample was fixed to an aluminium stub using double-sided conductive

carbon tape to a sampling tray and dusted with an inert gas. Samples were subsequently

sputter-coated with a mixture of gold:palladium (80:20) to form a layer of approximately 28 nm

on the surface of the samples. In order to investigate the internal morphology of the different

bead samples, one or more beads of each sample were cut in half with a scalpel under a

stereomicroscope and the internal structure of these beads were coated with the gold:palladium

coating (Marais et al., 2013:6742; Sungthongieen et al., 2004:149). An Eiko® ion coater

(model IB-2, Eiko Engineering, Tokyo, Japan) was used in all coating procedures and operated

under a vacuum higher than 0.06 Torr. A FEI Quanta® 250 Environmental Scanning Electron

Microscope with a Field Emission Gun (FEI©, Eindhoven, Netherlands) was used to study the

samples and displayed on a commercial computer (Frizon et al., 2013:534; Marais et al.,

2013:6742).

3.4.1.2 Particle size analysis

Particle size analysis offers essential information reflecting the mean particle size and particle

size distribution within a powder or bead formulation. Understanding these physical properties

of powders and beads enables the formulation scientist to explain observed behavioural

differences between powders, especially in terms of powder flowability (Horn, 2008:38).

Particle size analysis of the cassava starch samples was conducted with a

Malvern® Mastersizer® 2000 instrument fitted with a Hydro 2000SM small volume dispersion unit

(Malvern® Instruments, Malvern, UK). The Hydro 2000SM dispersion unit was employed during

the particle size analysis of the raw material samples. For particle size determination of the

bead formulations, the analysis was performed with a 2000MU dispersion unit fitted to the

Mastersizer® instrument. As dispersion medium for all samples (powder and bead samples),

absolute ethanol was used at a stirring rate of 1500 rpm. The small volume dispersion unit

(Hydro 2000SM) was filled with 100 ml absolute ethanol for the powder samples, whereas the

(Hydro 2000MU) dispersion unit for the bead formulations was filled with 500 ml absolute

ethanol. A background measurement was taken for all samples to compensate for electrical

interference as well as possible interference from the dispersion medium. Upon completion of

the background measurement, a sample of the appropriate material was added to the

dispersion unit. Samples of the beads were dispersed in 6 ml absolute ethanol prior to addition

Page 53: Cassava starch as modified release excipient in selected

52

to the small volume dispersion unit. A sufficient quantity of the sample was added to obtain an

obscuration of between 10 and 20%. After a suitable obscuration was obtained, the particle

size of each of the samples was measured. Each measurement consisted of 12000 sweeps.

The particle size and distribution of each sample were measured in triplicate and calculated with

Malvern® Software (Malvern® Instruments, Malvern, UK).

3.5 FLOW PROPERTIES

Flow performance is often best described by quantification of the flow process. Several

methods have been defined, either directly, using dynamic or kinetic methods, or indirectly,

normally by measurements conducted on static powder beds (Staniforth, 2002:601). This

section describes the different methods utilised to determine the flow properties of the various

starch samples and bead formulations. These include the critical orifice diameter, flow rate,

angle of repose, powder density and compressibility.

3.5.1 CRITICAL ORIFICE DIAMETER

Critical orifice diameter (COD) is defined as the smallest orifice through which a powder will flow

freely without the application of any external aid or interference. The apparatus, developed by

Buys and co-workers (2005:40-42) was used to determine the COD of the powders (figure 3.1).

A set of copper rings (between 5 and 10 mm thick) with a centrally located orifice was used to

determine the critical orifice diameter. By placing the copper rings in increasing size on top of

one another, a tapered cone was formed. Each ring has a different size opening and the orifice

of each disc was machined to a set angle. The largest disc opening was 32 mm and the

smallest was 1.5 mm. A stainless steel hopper was fitted to the top of the funnel to create a

holding chamber for the powder. This set was placed on top of a three legged stand to a height

of 95 mm.

Page 54: Cassava starch as modified release excipient in selected

53

Figure 3.1: Apparatus used for the critical orifice diameter determination

A powder or bead formulation mass of 100 g was gently poured into the holding cylinder, while

the opening on the bottom ring was kept shut. Opening the bottom orifice resulted in the

discharge of the powder or bead formulation (if possible) from the holding chamber.

Interchanging the stacked rings allowed for changing the bottom orifice diameter, whilst keeping

the slope of the funnel constant until the smallest diameter was found through which each

powder could flow freely. This specific diameter was noted as the COD. Each study for both

original and dried powder, as well as for each bead formulation, was done in triplicate and the

average COD, standard deviation (SD) and percentage relative standard deviations (%RSD)

were calculated (Buys, 2005:40-42; Lambrechts, 2008:40).

3.5.2 FLOW RATE

The most direct method of assessing powder flow properties is the hopper flow rate. This

method describes the amount of powder that could be discharged through a funnel in a specific

time unit; normally per second (de Kock, 2005:64).

In order to determine the flow rate of the powders (original and dried samples) and bead

formulations, a stainless steel hopper with a diameter of 30 mm was used. A hopper, fitted with

a closed shutter at the bottom and which was raised 100 mm above the work surface, was filled

with a predetermined amount of powder or beads (approximately 100 g).

Subsequently, the shutter was opened and the time required to complete the discharge of the

powder mass was recorded (Lavoie et al., 2002:887-893). By dividing the powder weight with

Page 55: Cassava starch as modified release excipient in selected

54

the time recorded, a flow rate for the specific sample was calculated (using either equation 3.3

or 3.4). The procedure was performed in triplicate using different samples (100 g each) and the

average flow rate (g.sec-1), SD and %RSD were calculated (Sonnekus, 2008:23).

T

MF [3.3]

T

VF [3.4]

Where F represents the flow rate in g.sec-1; t represents time (sec). Mass (in g) is represented

as M and volume (in ml) as V.

3.5.4 ANGLE OF REPOSE

Angle of repose (AoR) is defined as a dynamic and static angle at which a powder comes to rest

when discharged from a container (Geldart, et al., 2006:104). Interactions between cohesive

and free-flowing powders can influence the flowability of powders. A higher angle indicates a

greater cohesive powder, whereas a lower angle is suggestive of a less cohesive powder

(Staniforth et al., 2007:170; BP, 2015: XVII N). The following table (table 3.3) provides the

quality of flow respective to possible angles at which a powder may come to rest.

Table 3.3: Flow quality of powders for various angles of repose (revised from Wells et al.,

2007:356)

Flow quality Angle of repose (degrees)

Excellent < 20

Good 20 - 30

Acceptable 30 - 34

Very poor > 40

After the complete discharge of the powder and bead formulations from the hopper, the height

and diameter of the resulting heap were measured and recorded (Martin et al., 1993:447;

Staniforth, 2002:207; Wong, 2002:2636). Figure 3.2 below represents these factors and their

respective dimensions in regard to the resting heap. Each experiment was done in triplicate.

Using the obtained data, the angle of repose was calculated using the following equation:

Page 56: Cassava starch as modified release excipient in selected

55

r

hTanθ [3.5]

Where h represents the height (mm) of the powder cone and r (mm) is the radius of the base

cone.

Figure 3.2: Angle of repose of a resting powder heap (bed)

3.5.4 POWDER DENSITY

Another characteristic of powders that influence powder flow which merits consideration, is the

density of a powder or formulation. The density of matter, including powders and beads, is

described as the mass of that matter divided by the volume that amount of matter may displace.

The flow of powder is affected by the density of a powder. Another property directly influenced

by the density of powders, is the compressibility of a powder. More densely powders tend to

have a weaker flowability, whereas a less dense powder tends to flow more freely (Jallo et al.,

2012:213; Traina et al., 2013:843).

The bulk and tapped densities of the powders and bead formulations were determined by

pouring 100 g of cassava starch or bead formulation into a graduated measuring cylinder. The

initial occupied volume was measured and the filled cylinder was placed on an

Erweka® Tapped Density Tester SVM 12/221 (Heusenstamm, Germany), which was set at an

amplitude of 5 A. Each sample was vibrated until a constant volume was obtained (BP,

2015:XVII A). Powder densities were calculated by the following equations:

b

bV

mρ [3.6]

Page 57: Cassava starch as modified release excipient in selected

56

p

pV

mρ [3.7]

t

tV

mρ [3.8]

Bulk density (ρb) was calculated as the ratio of the mass (m) to the initial (bulk) volume (Vb).

Similarly, the tapped density (ρt) was calculated as the ratio of mass to the final (tapped volume,

Vt) volume of the sample.

3.5.5 COMPRESSIBILITY

Carr’s index (percentage compressibility) and the Hausner ratio were respectively calculated

from the calculated powder and bead formulation densities. This provided a better

understanding of the compressibility of the starch powders and bead formulations

(BP, 201:XVII N, Jallo et al., 2012:216; Traina et al., 2013:843). Table 3.4 reflects the flow

quality corresponding to different values of Carr’s index and Hausner’s ratio. Equation 3.9 and

3.10, were employed to determine the compressibility.

t

bt

ρ

ρρ(CI) Index sCarr'

[3.9]

b

t

ρ

ρRatio Hausner [3.10]

Table 3.4: Flow quality as indicated by Carr’s index and the Hausner ratio (revised from

Aulton & Wells, 2002:134)

Flow Quality Carr’s Index (%) Hausner Ratio

Excellent (free flowing) 5 - 15 1.05 - 1.18

Good 15 - 18 1.18 - 1.22

Fair 18 - 21 1.22 - 1.27

Acceptable 23 - 28 1.27 - 1.39

Poor 28 - 35 1.39 - 1.54

Very poor 35 - 38 1.54 - 1.61

Page 58: Cassava starch as modified release excipient in selected

57

Extremely poor (cohesive) > 40 > 1.61

3.6 EVALUATION OF THE BEAD FORMULATIONS

3.6.1 FRIABILITY

Durability is another consideration of importance for all SODFs. Mechanical and physical

resilience are required during transport and handling. In order to determine the durability of

SODFs, the friability was determined. This can be simulated by a tumbling motion of a SODF in

a friabilator (Allen et al., 2011:233).

Friability was measured using an Erweka® Friabilator (Type TAR 220, Heusenstamm,

Germany). As described by the BP (2015), a bead sample of approximately 3 g from each

formulation, which was dusted and weighed beforehand, was loaded into the friabilator; 10 glass

beads were added to this sample. The initial weight was recorded as W0. The apparatus was

run for a total of 100 revolutions; at 25 rpm for 4 min, followed by the removal of the sample.

These samples were dusted and weighed again (W1) (BP, 2015:XVII G), and the percentage

friability was calculated for each formulation using equation 3.11. Each sample was evaluated

in triplicate.

x100W

WWFriability %

0

10 [3.11]

3.6.2 SWELLING AND MASS LOSS

Swelling was evaluated per published method (Singh et al., 2009:1123). A bead sample of

approximately 250 mg was evaluated for each formulation where the initial weight (W0) was

recorded beforehand. Each sample was placed in a basket and introduced into a USP type II

dissolution apparatus at 37 ± 0.5°C. The dissolution medium used was 675 ml of a 0.1 M

hydrochloric acid (HCl) solution for the first 2 h. Subsequently 225 ml of a 0.2 M phosphate

buffer was added and the pH adjusted to 6.8. Samples were drawn at predetermined time

intervals of 30, 60, 90, 120, 180, 360, 480, 600 and 720 min, blotted with filter paper and

weighed. The measurement of the swollen weight (W1) was noted. In order to determine the

loss (erosion) of the matrix, the swollen samples were dried in a regulated oven at 40 ± 05°C for

12 h and weighed afterwards to record the mass after erosion (W2). Percentage swelling as

well as percentage erosion was calculated with the following equations:

Page 59: Cassava starch as modified release excipient in selected

58

x100W

WSwelling %

0

1 [3.12]

0

21

W

xWWErosion % [3.13]

3.6.3 DISINTEGRATION

Disintegration of SODFs is recognised by the loss of its initial size as a result of the initial unit

breaking into smaller pieces. As described in chapter 2, the SODF is fragmented into smaller

particles; this in turn increases the surface area of the unit, thereby increasing the rate of

dissolution. Disintegration is a constant process (Ashford, 2007:300).

Empty capsules (size 0) were weighed. Each capsule was subsequently filled with beads and

weighed again. Six capsules containing spheronised beads were evaluated for disintegration.

This was evaluated using a disintegration tester (Erweka® Type ZT 323, Heusenstamm,

Germany). Distilled water was used as the disintegration medium and maintained at 37 ± 0.5°C

with a thermostat. The encapsulated beads were placed in baskets attached to the

disintegration tester, these baskets were dropped into the medium and then raised out of the

medium and this process was repeated until the capsules dissolved. The time it took for each

capsule to disintegrate was recorded (BP, 2015: XVII A).

3.6.4 ULTRAVIOLET-SPECTROPHOTOMETRIC ANALYSIS

Analytical chemistry is the field which encompasses the many methods relevant to chemical

analysis and quantification. Various methods can be employed in order to determine the

chemical composition or chemical presence of unknown substances, these include:

spectrometric, titrimetric and chromatographic methods (Krull et al., 2014:1-7). In the

pharmaceutical industry it is vital to determine the quality, composition and quantities of the drug

and other excipients present in dosage forms. One of the most widely used methods is

ultraviolet-spectrophotometry (UV-spectrophotometry). The science of UV-spectrophotometry is

based on the analysis of the energy transition that occurs when a compound is irradiated with

ultraviolet light. UV-spectrophotometry is a cost-effective analytical method which can be

utilised in order to determine the presence of a compound in solution, if a reference for

comparison for that specific compound is available. This rational can be used to determine the

concentration of a drug in solution, which in turn can be used to determine the dissolution

Page 60: Cassava starch as modified release excipient in selected

59

behaviour of a drug from a dosage form (Kassab et al., 2010:968-971, Krull et al., 2014:10;

Wilson et al., 2005:591-599).

3.6.4.1 Standard curve

A 25 mg gliclazide sample was vortexed in 10 ml methanol. The solution was added to 75 ml

methanol and placed in an ultrasonic bath (Labotec© EcoBath® model 103, Labotec©, Midrand,

South Africa) for 20 min. A standard solution of 250 ml was made by adding a 0.1 M

HCl-solution to the methanol mixture, which in turn was ultrasonicated for 20 min. The solution

was filtered with a 0.45 µm nylon membrane pre-filter attached to a syringe in order to remove

contaminants. The resulting filtrate wasultrasonicated for another 20 min. A concentration

range of 2 - 40 µg.ml-1 was prepared. These concentrations were acquired by adding 5 ml of

the stock solution to a 250 ml volumetric flask; 10 ml to a 100 ml flaks; 20 ml to a 100 ml flask;

15 ml to a 50 ml flask and 20 ml to a 50 ml flask. Each sample was made up to volume with

deionised water. A spectral analysis at 229 nm was conducted using an Analytikjena® UV-

spectrophotometer (Speccord® 200 Plus, Jena, Germany). The absorbance values obtained

from these analyses where used to determine if a linear relationship exists between the various

concentrations within the range. In order to determine the precision of this method, inter- and

intraday validations were conducted (Kassab et al., 2010:986-971, Jamadar et al., 2011:339).

3.6.4.1.1 Interday precision

Using the method described in 3.6.4.1 an analysis for linear regression was conducted in

triplicate on the same day to determine the precision of the range used, with a resulting %RSD

of less than 5% (Chinyemba, 2012: 27-29; Marais, 2013:98-101).

3.6.4.1.2 Intraday precision

For intraday variance the method described above was repeated on three consequent days.

%RSD must be less than 5% (Chinyemba, 2012: 27-29; Marais, 2013:98-101).

3.6.5 DISSOLUTION BEHAVIOUR

The dissolution behaviour of pharmaceutical dosage forms is important with regards to

determining the release profile of the drug from the dosage form. By comparing the dissolution

behaviour and release of a commercial SODF (Diamicron®) to that of an experimental SODF, it

is possible to determine the viability of the experimental SODF as a candidate for modified

release SODFs.

Page 61: Cassava starch as modified release excipient in selected

60

3.6.5.1 Assay

In order to determine the drug loading capacity of the beads, a 100 mg bead sample from each

formulation was crushed using a mortar and pestle. It was dispersed in 100 ml ethanol, stirred

for 12 h and sonicated in a Labotec EcoBath® (Model 103, Labotec©, South Africa) for 30 min.

The subsequent suspension was filtered through a 0.45 µm membrane filter. A 3 ml sample

was pipetted into a 100 ml volumetric flask to obtain the correct dilution. This dilution was

analysed at a wavelength of 229 nm using a spectrophotometer (Speccord 200 Plus,

Analytikjena®, Germany) (Chinyemba, 2012:45).

3.6.5.2 Dissolution studies

Dissolution studies were conducted using a USP paddle method in a six station dissolution

apparatus (Distek® 2500 dissolution apparatus, USA). For the first two hours 675 ml of

0.1 M HCl was used as the dissolution medium. Thereafter the pH was adjusted to pH 6.8 by

adding 225 ml phosphate buffer (pH 6.8). The stirring rate was set at 50 rpm and the

temperature maintained at 37 ± 0.5°C. Samples of approximately 5 ml were withdrawn using an

auto sampler (Distek® evolution 4300, USA) at predetermined time intervals of 0, 2.5, 5, 7.5, 15,

30, 60, 90, 120, 180, 240, 360, 480, 600, 720 and 1440 min. A sample was withdrawn at the

24 h interval; the stirring rate was adjusted to 250 rpm for a further 15 min and the last sample

was collected (BP, 2015:XII B; Singh et al., 2009:1123; USP, 2008:268-269). Samples of 3 ml

withdrawn at the various time intervals were diluted to a volume of 10 ml. The withdrawn

volume from the vessels, were replaced. This replacement medium was collected from a vessel

containing blank medium which was calibrated at each pH level and kept at the same

temperature as the experimental vessels. All withdrawn samples were analysed with a ultra-

violet (UV) spectrophotometer at 229 nm (BP, 2015: XII B; Singh et al., 2009:1123; USP,

2008:268-269).

3.7 STATISTICAL ANALYSIS

To compare the dissolution profiles, three statistical parameters were calculated. These were

the mean dissolution time (MDT), the dissimilarity factor (f1) and the similarity factor (f2). MDT is

the statistical moment of the cumulative dissolution process and is the mean time taken for the

drug to dissolve under in vitro conditions (Reppas & Nicolaides, 2000:231-232). MDT was

calculated with the following equation:

Page 62: Cassava starch as modified release excipient in selected

61

n

1i d

n

1i dmid

Δx

ΔxtMDT [3.14]

Where, MDT is the mean dissolution time in minutes, i the sample number, n the total number of

sampling times, tmid the midpoint between i and i-1 and Δxd the additional mass dissolved

between i and i-1 (Chinyemba, 2012:29; Marais, 2013:54-56)

Moore and Flanner (1996:64-74) used the similarity factor (f2) to compare dissolution profiles.

This factor compared the difference between the percentage drug dissolved per unit time for a

test and reference formulation. The value of the similarity factor is 100 when two dissolution

profiles are identical and approaches 0 as the dissimilarity increases. According to the

Food and Drug Administration (FDA), two dissolution profiles can be considered similar when f2

values between 50 and 100 were obtained (Costa et al., 2001:129). The following equations

can be used to calculate the dissimilarity factor (f1) and the similarity factor (f2)

100%

R

TR

fn

1t

t

n

1t

tt

1

[3.15]

100TRn

1150logf

0.52

1t

2

tt2

[3.16]

Where, f1 is the difference factor and f2, the similarity factor. Rt represents the assay time at

time t, Tt the test assay at the same time, n the number of pull points and wt the optional weight

factor.

3.8 SUMMARY

To determine the viability of this study, the previously described experimental methods were

utilised. It was the hope that through these methods a suitable formulation for modified release

could be produced in order to provide a modified release dosage form. The experimental

methodology was employed in such a manner to optimise a formulation containing suitable

excipients and concentrations of all the relevant ingredients, including the drug. The viability of

cassava starch as an excipient in modified release dosage forms was also determined using

Page 63: Cassava starch as modified release excipient in selected

62

these methods. In chapter 4 the results of these experimental methods will be given and

discussed in order to provide a clearly defined picture.

Page 64: Cassava starch as modified release excipient in selected

63

Chapter 4

EXPERIMENTAL RESULTS

4.1 INTRODUCTION

The determination of flow properties provides valuable information on powder mixtures intended

for the manufacturing of SODFs. Arguably, the most important properties that influence powder

flow are the size and morphology of powder particles. A range of parameters or properties can

be investigated to determine whether a powder exhibits acceptable flow. These parameters or

properties include compressibility, flow rate, angle of repose and critical orifice diameter, to

name a few. Another factor which could affect the quality of powder flow is the moisture content

of the powder. Moisture present in the powder can significantly diminish a powder’s flow quality

(Emery et al., 2009:409).

Physical characteristics of SODFs influence a product’s commercial suitability. These

characteristics include: friability, disintegration time and dissolution profile of a specific SODF.

Friability provides an indication of the robustness of the product during transport and handling.

Disintegration describes the process by which an SODF is broken down into smaller pieces and

as a consequence increase the surface area available for drug dissolution. Disintegration time,

therefore, is the time taken by the SODF to break up into smaller particles as specified by the

official pharmacopoeias (BP, 2015:XII A1). Dissolution profiles are used to determine the rate

and extent of drug release from a dosage form. Furthermore, dissolution data is useful in

determining the similarity or difference between respective formulations. Dissolution data may

also be employed to determine whether the release of a drug from a particular dosage form is

conventional or modified (Lourenҫo et al., 2013:367-368).

The different formulation variables and their levels were investigated by means of a fractional

factorial design (as discussed in Chapter 3). In order to provide a simplified method of

reference to the different formulations, it was decided to provide an identifier, as seen in

table 4.1. The identifier consists of a sequence of letters and numbers, representing excipients

and levels, for example, M.G5.5.5; where M/C is the filler; G5 is the drug and its concentration

(% w/w); the second 5 is the concentration (% w/w) of the binder and the final number (5)

represents the concentration (% w/w) of HPMC in the formulation.

Page 65: Cassava starch as modified release excipient in selected

64

Table 4.1 Identifiers for each successful formulation and the composition of each formulation

Identifier

Filler Gliclazide

concentration (% w/w)

Kollidon® 30 concentration

(% w/w)

HPMC concentration

(% w/w) Type Concentration

(%)

M.G5.3.5* Avicel® 87 5 3 5

M.G5.5.10 Avicel® 80 5 5 10

M.G10.5.5 Avicel® 80 10 5 5

M.G10.0.10 Avicel® 80 10 0 10

M.G15.0.5 Avicel® 80 15 0 5

M.G15.3.10 Avicel® 72 15 3 10

C.G5.5.5 Cassava 85 5 5 5

*M/C = Filler (M = Avicel® or C = Cassava), G5 = concentration (% w/w) of gliclazide, second

number (3) = concentration (% w/w) of Kollidon® 30, last number (5) = concentration (% w/w) of

HPMC

4.2 PHYSICAL CHARACTERISTICS OF CASSAVA

STARCH

Selected physicochemical properties of the cassava starches were evaluated accordingly to the

methodology as put forth in chapter 3, in order to decide which type of starch w could be used

as received or whether further processing was necessary. These properties tested included

relative humidity (RH) and moisture content. Infrared (IR)-spectrometry was employed to

determine if the two samples of starch were identical or represented different crystal forms of

the starch.

4.2.1 MOISTURE CONTENT AND THERMAL ANALYSIS

The average moisture content as determined by means of Karl-Fischer titrations of the

purchased and donated cassava starch is presented in figure 4.1. At time 0 min, the moisture

content for each sample was 15.82 ± 0.339% and 11.84 ± 0.156%, respectively.

Page 66: Cassava starch as modified release excipient in selected

65

4

6

8

10

12

14

16

0 30 60 120 240 360 480

Ave

% H

2O

Co

nte

nt

Time (min)

Donated

Purchased

Figure 4.1: Average moisture content of the donated and purchased Cassava starch,

at 40°C for various drying times

Powders that contain high levels (> 10%) of moisture have been associated with poor flowability

as well as poor powder characteristics (Crouter & Briens, 2014:70-73; Emery et al. 2009:414;

Nokhodchi, 2005:50). In order to determine whether the moisture was due to hygroscopicity or

a constituent of the polymer matrix, differential thermal (DT) and thermogravemetric (TG)

analyses were conducted. The thermograms are depicted in figures 4.2 and 4.3.

From these thermograms it was evident that weight loss, due to moisture evaporation, occurred

from the initial onset of heating, therefore indicating that the moisture present in the starch

samples was not part of the polymer matrix itself, but was present in the powder due to

hygroscopicity. Consequently, the temperature and time interval at which the moisture content

would be acceptable (6 – 10%) to provide conditions at which the flowability of the powder

would be acceptable, was determined (Crouter & Briens, 2014:70-73; Emery et al. 2009:414;

Nokhodchi, 2005:50).

After drying the starch samples at 25°C, 30°C and 40°C, over a time period of eight hours, the

moisture content was re-evaluated. It was determined that for the samples to show acceptable

flow, it should be dried at 40°C for 4h in order to achieve a moisture content of 6 – 10%. The

average moisture content of the samples dried at 40°C for 4h was measured at 8.07 ± 0.007%

and 8.79 ± 0.389%, respectively (Viljoen et al., 2014:730-742).

Page 67: Cassava starch as modified release excipient in selected

66

Figure 4.2: Thermogram of donated Cassava starch

Figure 4.3: Thermogram of purchased Cassava starch

Page 68: Cassava starch as modified release excipient in selected

67

4.2.2 INFRARED-SPECTROSCOPY

Figures 4.4 and 4.5 reflect the IR-spectra for both the donated and the purchased starch.

Figure 4.4: Overlay of IR-spectra for the donated (red) and purchased starch (black)

According to the IR-spectrum region of 1500 – 350 cm-1, the two samples of cassava starch

proved to be similar. This area serves as a fingerprint to determine, identify and compare

substances. Both samples contained a high amount of OH-groups and strong N-triple bonds.

In order to confirm the composition of the starch samples, it was opted to improve the resolution

of the IR-spectra. The resolution of the spectra was improved with the use of a Fourier

transform IR-spectrometry (FTIR). This improved resolution can be seen in figure 4.5.

Figure 4.5: IR-spectra from FTIR-analysis of the donated and purchased Cassava starch

Page 69: Cassava starch as modified release excipient in selected

68

FTIR provided a higher resolution and identification of the finger print region,

1500 – 350 cm-1, which indicated that the two starch samples were indeed related. However,

the improved resolution did indicate differences between the two samples. This proved that

both samples were of two unique starches (Vicentini et al., 2007:756-758). The finger print

region of the IR-spectra (obtained using FTIR-analysis) correlated with the IR-spectra given by

Huang et al. (2007:133).

4.3 PRELIMINARY EXPERIMENTS AND BEAD

MANUFACTURING

A 100 mg sample of purchased cassava starch was wetted with distilled water to determine if a

wet mass suitable for extrusion could be formed. The purchased starch was selected due to its

lower moisture content and possibly higher flowability. This mass needed to have a firm

consistency in order to be introduced into the extruder to obtain an acceptable extrudate. These

extrusions would be used in the manufacturing of beads. The first attempt proved difficult in

producing a mass of acceptable consistency for extrusion. A wet mass was produced with a

high concentration of water. This mass seeped through the perforations of the extrusion screen.

A mass of this consistency proved inefficient for the production of beads. Consequently, it was

decided to use a different wetting agent. An ethanol-water mixture was selected for wetting the

mass. With the addition of this wetting agent to another 100 g sample, a firmer wetted mass

was produced. This improvement in the consistency of the wetted mass correlates with the data

acquired by Millili and Schwartz., (1990:1411) who found that an ethanol:water mixture provided

a firmer consistency for extrusion-spheronisation. At 32 rpm, the radial extruder produced

extrusions of adequate consistency for spheronisation. After repeated adjustments with regards

to the rotation speed of the spheroniser as well as the duration of spheronisation, spherical

beads with irregular surface characteristics were obtained (Dhandapani et al., 2012:10-16; Joshi

et al., 2011:113). Kumar et al. (2012:1) stated that ideally beads should have a spherical shape

and a size range of approximately 600 – 1000 µm.

To improve bead quality with regards to size and shape, Kollidon® VA64 was added as binder.

The inclusion of Kollidon® VA64 was based on its use in matrix and tablet formulations

(Bhaskaran & Lakshmi, 2010:2431; Bühler, 2008:199-241). However, the mass that was

produced depicted a more viscous consistency which made extrusion difficult. It was decided to

investigate the the substitution of Kollidon® VA 64 with Kollidon , as an alternative binder.

Kollidon® 30 provided a mass with a less viscous nature; and consequently the extrusion-

Page 70: Cassava starch as modified release excipient in selected

69

spheronisation of the beads was successful. Beads produced from Kollidon® 30-containing

mixtures depicted a more spherical shape. The production of spherical beads prompted the

addition of gliclazide. A fractional factorial design (Table 3.2) was employed to investigate the

effects of excipients and concentrations on bead formulation.

Microcrystalline cellulose (Avicel®)was selected as alternative filler to cassava starch. Avicel®

is an industry standard for both direct compression and bead production (Dukic-Ott et al.,

2009:38-39; Vervaet et al., 2008:39). Smooth and spherical beads were successfully produced

with Avicel® containing formulations. After this production it was opted to improve the bead

spherocity by adding HPMC. HPMC was also selected for its application in matrix based

SODFs. However, Avicel® formulations containing no HPMC tended to produce irregularly

shaped and non-spherical beads, whereas all formulations containing HPMC rendered spherical

beads. HPMC has been described by Gandhi et al. (1999:166-168) as a recommended aid with

regard to bead manufacturing and bead quality. This could be attributed to the water solubility

and consequent gelling of HPMC, which is a low molecular weight polymer (Dukiƈ-Ott et al.,

2009:42-43; Gandhi et al., 1999:166-168). Cassava starch on the other hand, produced one

viable formulation that consisted of spherical beads. These beads therefore had a desirable

shape, size and mechanical strength. The success of the formulation, C.G5.5.5, could be

attributed to the respective concentrations of each excipient and the drug (Gandhi et al.,

1999:163-165; Khan et al., 2001, 350-354; Vervaet et al., 1995:136-143).

The aforementioned process in conjunction with the factorial design was used to determine

which mixtures would provide acceptable beads for the remainder of this study. These

formulations with acceptable quality is identifiable in table 4.1.

Table 4.2 provides the selected formulations and an indication of whether a successful

formulation could be manufactured from the selected combination of excipients.

Page 71: Cassava starch as modified release excipient in selected

Table 4.2: Selected formulations and respective excipients and concentration

Identifier Type of filler Filler

concentration

Drug

concentration

Kollidon

concentration

HPMC

concentration

Experimental

status*

M.G5.00 Avicel 95 5 0 0 Unsuccessful

C.G5.30 Cassava 92 5 3 0 Unsuccessful

M.G5.3.5 Avicel 87 5 3 5 Successful

C.G5.5.5 Cassava 85 5 5 5 Successful

M.G5.5.10 Avicel 80 5 5 10 Successful

C.G5.0.10 Cassava 85 5 0 10 Unsuccessful

M.G10.3.0 Avicel 87 10 3 0 Unsuccessful

C.G10.3.0 Cassava 86 10 3 0 Unsuccessful

M.G10.5.5 Avicel 80 10 5 5 Successful

C.G10.0.5 Cassava 85 10 0 5 Unsuccessful

M.G10.0.10 Avicel 80 10 0 10 Successful

C.G10.3.10 Cassava 77 10 3 10 Unsuccessful

M.G15.5.0 Avicel 80 15 5 0 Unsuccessful

Page 72: Cassava starch as modified release excipient in selected

C.G15.0.0 Cassava 75 15 0 0 Unsuccessful

M.15.0.5 Avicel 80 15 0 5 Successful

C.G15.3.5 Cassava 77 15 3 5 Unsuccessful

M.G15.3.10 Avicel 72 15 3 10 Successful

C.G15.5.10 Cassava 70 15 5 10 Unsuccessful

Page 73: Cassava starch as modified release excipient in selected

72

4.4 MORPHOLOGY AND SIZE

4.4.1 MORPHOLOGY

The morphology of the cassava starch powder particles was visualised by means of SEM, as

described in section 3.4.1.1 of this study. Both purchased and donated powders exhibited

spherical particles with occasional surface irregularities due to indentations (figure 4.6). From

these images it could be seen that both starch samples were in the same size range. It was

furthermore clear that the majority of the particles for both starches were less than 50 µm. As

poor flowability is usually observed for powders with an average particle size smaller than

100 µm, it would be expected that both starches will probably exhibit poor powder flow.

Additionally, agglomeration behaviour was observed in the micrograph depicting the particles of

the donated starch. Agglomeration behaviour is usually evident for small particles, indicating

cohesive behaviour which affects powder flow negatively (Kim et al. 2005:182-186; Landillon

et al., 2008:178-179, Lavanya et al., 2011:1338-1339; Staniforth & Aulton, 2007:169).

A B

Figure 4.6: Scanning electron microscopy micrographs of (A) purchased and (B) donated

starch

In figure 4.7 SEM-micrographs from the individual bead formulations are shown. For each

formulation, the bead shape, surface and internal structure are demonstrated, as indicated by

the letters A, B and C, respectively. Each micrograph in each (A, B and C) was conducted on

the same scales of magnification respective to that set. Set A had a scale of 1:500 µm, B a

scale of 1:20 µm and C a scale of 1:10 µm.

Page 74: Cassava starch as modified release excipient in selected

M.G5.3.5 M.G5.5.10 M.G10.5.5 M.G10.0.10 M.G15.0.5 M.G15.3.10 C.G5.5.5

A

B

C

Figure 4.7: SEM - micrographs of the different bead formulations (each set of three micrographs represents the following: A - full

view of the beads, B - the exterior surface morphology and C - the internal structure)

Page 75: Cassava starch as modified release excipient in selected

74

It was evident from figure 4.7 that a fairly spherical morphology was exhibited by the majority of

the formulations. The formulations, M.G5.3.5 and C.G5.5.5, each depicted a more spherical

shape whereas the remaining formulations portrayed either a bean (M.G10.5.5) or dumbbell

(M.G15.3.10) shapes that were indicative of insufficient spheronisation (Koester & Thommes,

2010: 1549-1550; Vervaert et al., 1995:136-141). These findings are in agreement with Chopra

et al. (2013:139), who stated irregularities in shape could occur with Avicel®-containing

formulations. The exterior surface morphology of each Avicel® formulation (figure 4.7 A) was

smooth and could be suggestive of good flowability (Kim et al. 2005:182-186; Lavanya et al.,

2011:1338-1339).

Avicel® formulation C.G5.5.5 depicted a general smooth surface, however, with closer

inspection of the exterior morphology (figure 4.7 B); a rough surface could be observed. This

formulation was the only formulation that contained cassava starch and considering the

micrograph of the cassava starch, the rough surface might have been attributed to the cassava

particles. The individual beads were grouped together with the assistance of a web-like matrix.

This matrix was formed due to the addition of the binder, HPMC. Both HPMC and Kollidon®

tend to form polymer matrices which influence the binding of excipients, as well as drug release

(Budiasih et al., 2014:54; Ingle et al., 2013:13; Mustafa et al., 2014:309-311).

In contrast to the cassava-containing formulation (C.G5.5.5), the individual particles of the

Avicel® filler could not be identified (figure 4.7 B) in the Avicel® bead formulations. The internal

structure of the Avicel®-containing beads (figure 4.7 C) tended to be more densely packed

together, whereas, C.G5.5.5 clearly showed cavities within the beads. These voids or cavities

could encourage moisture to penetrate the interior core of the beads, which possibly influenced

the dissolution rate of the drug (Yang et al., 2014:187-196).

Page 76: Cassava starch as modified release excipient in selected

75

4.4.2 SIZE DISTRIBUTION OF POWDER PARTICLES

Figure 4.8 represents the size-distribution of the individual powder particles and

beads.

0

150

300

450

600

750

900

1050

1200

1350

1500

1650

Size

(u

M)

Samples

d(0.1)

d(0.5)

d(0.9)

Figure 4.8: Size distribution histogram of both starches and bead formulations. Where

d(0.1) = 10% of particles smaller than, d(0.5) = 50% particles smaller than, and

d(0.9) = 90% of particles smaller than

Upon comparison of the particle size of both starch powders it could be postulated that the

starch powders are likely to exhibit poor powder flow. Lui et al (2008:109) stated that if the

mean particle size of material is smaller than 100 µm, the cohesive forces between the particles

would be higher. This could negatively affect the flow of the powders (Hart, 2015:2; Liu et al.,

2008:109; Staniforth & Aulton, 2007:170). From the size distribution of the bead formulations it

can be seen that they depicted an average median size range (d(0.5)) of

800 – 1000 µm, as illustrated in figure 4.8. From the particle size data it is expected that all the

bead formulations should exhibit good or acceptable flow as the d(0.9) values for all the bead

formulations were > 1000 µm. This indicated that 90% of the beads in the measured samples

were larger than 1000 µm. Additionally, from figure 4.8 a clear size difference can be seen

between the cassava starch powder particles and the C.G5.5.5 beads. This difference in size

could presumably improve the flowability of the product (Hart, 2015:2; Lui et al., 2008:109;

Vervaet et al., 1994:131-132). Size variation of the beads could be attributed to variation that is

seen in all pharmaceutical manufacturing processes. These processes include the amount of

Page 77: Cassava starch as modified release excipient in selected

76

wetting agent added and the duration of time after extrusion, before introduction into the

spheroniser (Mallipeddi et al., 2010:56-62; 2014:362-366).

4.5 FLOW PROPERTIES

Several parameters were used to describe the flow behaviour of the cassava powders and the

bead formulations. These parameters and the values obtained are reported in table 4.3.

Both starches presented a critical orifice diameter (COD) value of 16 mm, whereas all the bead

formulations depicted a COD value of 6 – 7 mm. A high COD value indicates poor powder flow

and a smaller COD value is an indication of improved flow. The improved flow was a

consequence of the enlarged size of the beads as described in section 4.4. No marked

differences were observed pertaining to the COD values for the individual bead formulations.

Neither flow rate nor angle of repose could be determined for both starches. These results

indicated weak powder flow which corroborated the postulation in section 4.4, i.e. that the small

size of the starch powder particles is expected to be detrimental to powder flow. The moisture

content (15.82 and 11.84%) of the donated starch and purchased starch may also aggerasvate

the poor flow behaviour. Although a marked improvement (in comparison to the starch

powders) in the flow rate was observed for all bead formulations, differences could be seen

concerning the different bead formulations. These differences can be attributed to the shape of

each formulation’s individual beads as depicted in figure 4.6 and 4.7. As the shape of the

different bead formulations is not perfectly spherical, contact between the beads might increase

providing more friction and thus impeding flow (Javadzadeh et al., 2015: 86-97; Korhonen et al.,

2000:1141; Zhang et al., 2003:6-7; Zhang et al., 2004:371-390).

Page 78: Cassava starch as modified release excipient in selected

Table 4.3: Flow properties of both starches and bead formulations

Critical orifice diameter

(mm)

Flow Rate (g.s-1)

Average angle of

repose (°)

Density (g.cm-3)

Compressibility

Bulk Tapped Carr’s Index (%) Hausner Ratio

Donated 16 ± 0.0 No flow No flow 0.6 ± 0.01 0.8 ± 0.02 0.3 ± 0.01 1.4±0.02

Purchased 16 ± 0.0 No flow No flow 0.5 ± 0.02 0.8 ± 0.03 0.3 ± 0.01 1.5±0.02

M.G5.3.5 6 ± 0.6 4.8 ± 0.23 26.6 ± 0.22 0.8 ± 0.05 0.9 ± 0.01 0.1 ± 0.06 1.2±0.09

M.G5.5.10 6 ± 0.0 4.7 ± 0.13 28.3 ± 2.59 0.8 ± 0.01 0.9 ± 0.00 0.1 ± 0.02 1.1±0.02

M.G10.5.5 6 ± 0.0 4.6 ± 0.12 25.7 ± 1.45 0.8 ±0.01 0.9 ± 0.01 0.1 ± 0.01 1.1±0.01

M.G10.0.10 7 ± 0.6 5.2 ± 0.15 28.3 ± 1.23 0.8 ± 0.01 0.9 ± 0.00 0.1 ± 0.01 1.1±0.01

M.G15.0.5 6 ± 0.0 3.8 ± 0.08 29.8 ± 2.24 0.8 ± 0.00 0.9 ± 0.00 0.0 ± 0.01 1.0±0.01

M.G15.3.10 6 ± 0.0 3.6 ± 0.0 30.1 ± 0.22 0.8 ± 0.01 0.9 ± 0.00 0.1 ± 0.11 1.1±0.01

C.G5.5.5 6 ± 0.0 4.9 ± 0.14 26.3 ± 0.55 0.8 ± 0.01 0.9 ± 0.00 0.1 ± 0.00 1.1±0.01

Page 79: Cassava starch as modified release excipient in selected

78

Despite the differences in flow rate, all bead formulations exhibited an approximate flow rate of

3.64 – 5.17 g.sec-1, indicating good flowability. Both starches and all the bead formulations

exhibited Carr’s indices and Hausner ratios of < 1 and 1.07 – 1.18, respectively, which is

indicative of excellent (free) flow.

The bulk density of 0.60 g.cm-3 and 0.55 g.cm-3 for the donated and purchased starches,

respectfully, were less than that of the beads, which ranged from 0.79 - 0.83 g.cm-3. Tapped

densities of both starches and beads ranged from 0.81 – 0.91 g.cm-3. These results indicated

that the powders packed more densely after tapping, which could be ascribed to the smaller

particles, leaving less void spaces after being rearranged. The beads on the other hand, did not

rearrange as compact as the powder due to the rigidity of the beads and their inability to fill the

void spaces in-between. Considering the data for the different flow parameters, it is evident that

all the parameters on powder flow for the different bead formulations indicated good to excellent

flow behaviour. This is to be expected as beads are known to exhibit acceptable to excellent

flow due to their size and more spherical nature. It is therefore clear that the formulation of

beads resulted in a pronounced improvement in the flow properties in comparison to the

flowability of the starch powders.

From the data it could be concluded that cassava starch does not have an acceptable flow

quality, which excludes it as an excipient for direct compression without modification or the

inclusion of a glidant. However, the cassava bead formulation exhibited the necessary

flowability and therefore potential to be introduced into a tablet press for compression into a

multi-unit pellet system. The data also corroborates the influence of size on flow quality, by

increasing the size of the particles, with the help of bead manufacturing, the flow quality

improved dramatically.

4.6 EVALUATION OF BEAD FORMULATIONS

Each bead formulation was subjected to various experiments in order to determine the viability

of the individual formulations for manufacturing as a SODF. In this study one of the objectives

was to attempt the compression of the beads into a single unit product. The compression of

these beads into a single tablet is known as a multi-unit pellet system(Reddy et al., 2012:42-54).

This method of SODF manufacturing is based on the concept of providing a modified release

dosage form or a fixed dose combination. Each of these has their respective rationale in

support of multi-unit pellet system manufacturing. Different types of multi-unit pellet system

could be manufactured including direct compressed multi-unit pellet system and encapsulated

Page 80: Cassava starch as modified release excipient in selected

79

particulates. One of the most researched types, are manufactured with direct compression of

particulates into a single tablet (Reddy et al., 2012:42-54). The rationale behind a multi-

unit pellet system compressed from beads was in part to produce a convenient product for drug

delivery. Samples of the bead formulations were introduced into a Korsch® XP1 tablet press.

The tablets produced from these beads proved difficult to produce. As each produced batch

ejected from the die they either crumbled as they left the die or as they were moved . This

could be attributed to insufficient mechanical strength of the compressed tablets. The

insufficient mechanical strength may be attributed to the hardness of the individual beads being

too high for the necessary deformation in order to compress into a single tablet, or insufficient

cohesion between the individual beads. Therefore, it was decided to encapsulate intact beads

in size 0 gelatine capsules to render a SODF.

4.6.1 FRIABILITY

Friability of the individual bead formulations provided information regarding the resistance of the

beads to breaking or splitting during handling and transport (Chapter 3). Table 4.4 presents the

average percentage friability results obtained for the different bead formulations.

Table 4.4: Percentage friability of bead formulations

Bead formulations Average % friability

M.G5.3.5 1.6 ± 3.22

M.G5.5.10 0.1 ± 0.27

M.G10.5.5 0

M.G10.0.10 0.7 ± 0.20

M.G15.0.5 0

M.G15.3.10 0

C.G.5.5.5 2.0 ± 0.88

Friability values could only be successfully determined for four formulations as the other

formulations depicted extremely brittle beads. The analysis indicated that C.G5.5.5 provided a

poor friability (2.0 ± 0.88); and M.G5.5.10 and M.G10.0.10 passed the 1% acceptance value

specified by the British Pharmacopoeia (BP, 2015: XVII G).

Page 81: Cassava starch as modified release excipient in selected

80

4.6.2 SWELLING AND MASS LOSS

During the swelling and mass loss study the results relating to the cassava starch beads were

not obtainable due to the disintegration of the beads. With the removal of the basket containing

the cassava beads, the residue of these beads seeped out of the basket, which consequently

made the determination of the weight impossible. This could be ascribed to the pH susceptible

nature of cassava starch. Cassava rapidly dissolves in a low pH or acidic medium. Figure 4.9

reflects the cumulative increase/decrease in mass (mg) measured for the Avicel® bead samples

per time interval (min).

A drastic increase in mass was observed during the first 30 min for all of the Avicel® bead

formulations, whereafter no substantial increase or decrease in mass for the following 90 min

could be observed. During the first 120 min, the beads were exposed to an acidic medium

whereafter the medium was changed to a more alkaline medium (pH 6.8). In the alkaline

medium (180 – 720 min) clear differences could be observed between the different Avicel®

formulations.

Low gliclazide content allowed for a higher degree of swelling and this could be attributed to the

ease at which the moisture permeated the beads due to the fact that less hydrophobic drug was

present that would be able to form a barrier against water penetration into the beads. The

formulations containing a higher concentration of gliclazide (10 or 15% w/w) depicted decreased

swelling. The higher drug content might have decreased the rate as well as quantity of liquid

that penetrated the beads due to its natural hydrophobic character (chapter 2). Moreover,

formulations comprising higher concentrations of HPMC in combination with a lower

concentration Kollidon® 30 portrayed a marked increase in the percentage swelling. This

increased swelling could be attributed to the polymer rich HPMC. HPMC is highly hydrophilic

and thus attracts moisture into the beads resulting in swelling/expansion of the matrix. The

swelling of the polymer could cause pores present in the beads to open and allow more

moisture into the beads (Akhgari et al., 2007:51-58; Ghori et al., 2014:1-17;Scholtz et al.,

2014:486-501; Viridén et al., 20010:60-67; Viridén et al., 2011:470-479).

Page 82: Cassava starch as modified release excipient in selected

0

200

400

600

800

1000

1200

1400

1600

1800

0 60 120 180 240 300 360 420 480 540 600 660 720 780

Cu

mu

lati

ve

Ma

ss

In

cre

as

/De

cre

as

e (

mg

)

Time (min)

M.G5.3.5

M.G5.5.10

M.G10.5.5

M.G10.0.10

M.G15.0.5

M.G15.3.10

Figure 4.9: Cumulative mass increase or decrease of Avicel® beads as a function of time (min) after exposure to calibrated pH

environments.

Page 83: Cassava starch as modified release excipient in selected

82

According to Goyal et al. (2009:95-96) swelling of HPMCcontaining formulations are pH

dependent. This was also evident within this study as the amount of swelling in the alkaline

medium increased noticeably relative to the amount of swelling in the acidic medium. The lower

the concentration of Kollidon®, the lower the amount of swelling and also the mass loss. After

the swelling experiment, the beads were dried for 12 h until no noticeable weight loss was

observed. Annexure C displays the results obtained after drying of the beads and from these

results it was evident that the loss on mass of the different Avicel® bead formulations all

averaged approximately 170 mg after drying, thus portraying an approximate loss in mass of

30% (Akhgari et al., 2007:51-58; Ghori et al., 2014:1-17; Viridén et al., 2011:470-479).

4.6.3 DISINTEGRATION

All capsules, irrespective of formulation disintegrated in less than 5 min, which complies with

specifications of the British Pharmacopoeia (2015: XVII S). Upon disintegration of the capsule

shells, the beads were dispersed throughout the disintegration medium.

4.6.4 DISSOLUTION BEHAVIOUR AND STATISTICAL ANALYSES

Several variables needed to be optimised in order to evaluate the bead formulations. This

included the amount of beads required to compare dissolution profiles with the control product,

i.e. Diamicron®, and standardisation of the analytical method that was employed.

4.6.4.1 Standard curve

A stock solution was prepared according to the method described section 3.6.4. The solution

consisted of 25 mg gliclazide dissolved in a 2:3 methanol:HCl - solution. This stock solution

was used to prepare a standard solution to construct a standard curve which was utilised for the

first 2 h of the dissolution study. Another stock solution was prepared with a

2:3 methanol and phosphate solution with a pH of 6.8. This solution was used to prepare a

standard solution in order to construct a standard curve that was used for the remainder of the

dissolution study (2 hr to 12 hr).

4.6.4.2 Linearity

The following figure (figure 4.10) provided a graph depicting a linear relation between the

absorbance and concentration of gliclazide in a methanol:HCl solution.

Page 84: Cassava starch as modified release excipient in selected

83

y = 0.0373x - 0.0021R² = 0.9997

0.00

0.20

0.40

0.60

0.80

1.00

1.20

1.40

1.60

0 5 10 15 20 25 30 35 40 45

Ab

so

rba

nc

e (

un

its

)

Concentration (mcg.ml-1)

Figure 4.10: Standard curve for gliclazide dissolved in 2:3 methanol:HCl solution

Standard solutions were prepared according to the method described in section 3.6.5. Linearity

was observed in a concentration range of 2 – 40 µg.ml-1, with a R2 - value of ≥ 0.999 for both the

acidic and alkaline media. At concentrations higher than 40 µg.ml-1 deviation from Beer’s law

was observed. This phenomenon was also noted by Ibragimova and Ikramov (2015:73-74) with

another sulphonylurea, glieipiride. Glimepiride and gliclazide are both examples of the same

drug classification, namely second generation sulphonylureas, which are chemically and

pharmacologically related (Kalra & Gupta, 2015: 101-104, Kalra et al., 2015:314-315).

4.6.4.2.1 Intra- and interday precision

Intra- and interday precision fell within acceptable limits as a %RSD value of ≤ 5% was obtained

for both intra- and interday precision (Chinyemba, 2012: 27-29; Marais, 2013:98-101).

4.6.4.3 Dissolution

Dissolution studies were conducted on samples from each formulation containing 30 mg

gliclazide. The sample weight was determined based on assay results obtained as per

described method (section 3.6.5.1). The selection of the 30 mg drug dose per bead sample was

based on the drug dose of the control (Diamicron® tablets; 30 mg gliclazide per tablet). The

dissolution profiles of the different bead formulations as well as the control are presented in

figure 4.11.

From the dissolution profiles it is observed that the profiles of two formulations appear differently

from the other profiles, namely, formulation C.G5.5.5 and M.G15.3.10. From figure 4.11, it is

evident that C.G5.5.5 exhibited a burst release of drug within 15 min of the study.

Page 85: Cassava starch as modified release excipient in selected

0

20

40

60

80

100

120

0 60 120 180 240 300 360 420 480 540 600 660 720 780

Perc

en

tag

e D

isso

luti

on

(%

)

Time (min)

M.G5.3.5

C.G5.5.5

M.G5.5.10

M.G10.5.5

M.G10.0.10

M.G15.0.5

M.G15.3.10

Diamicron

Figure 4.11: Percentage of the drug dissolved as a function of time (min) within pH calibrated medium form simulating either a

acidic or alkaline gastric environments

Page 86: Cassava starch as modified release excipient in selected

85

This release is indicative of a fast release of the drug. Burst release can be advantageous in

certain applications e.g., drugs which require an immediate release and continuous release for

predetermined time interval, though in certain dosage form designs which require a slow initial

release, a burst release could be disadvantageous (Huang & Brazel, 2001:121-135). The burst

release may be attributed to the existence of cavities within the bead structure as was evident

from the SEM micrographs. It was postulated that the existence of these cavities might be

beneficial for liquid penetration into the bead structure and thereby benefited the dissolution rate

of the drug. This can contribute to the burst release seen within the first few minutes of the

dissolution study from this formulation. However, the remaining drug content (± 40 - 50%) was

released over the remaining period of the study. The corresponding release profile seen here

correlates with figure 2.2.

Although this formulation did not mimic the release profile of the control precisely, it is still

evident that extended release was observed over a period of approximately 6 h. Formulation

M.G15.3.10 exhibited a dissolution profile similar to the control, Diamicron®, under the test

conditions which indicated that it was possible to prepare a multiple pellet system that rendered

modified release of gliclazide.

Table 4.5: Mean dissolution time and similarity factor values for each bead formulation and

Diamicron®

Formulation Mean Dissolution Time

(min) Similarity Factor (f2)

M.G5.3.5 307.1 ± 27.17 37.3 ± 3.55

M.G5.5.10 201.7 ± 5.34 45.8 ± 3.11

M.G10.5.5 226.9 ± 9.21 49.9 ± 3.76

M.G10.0.10 268.3 ± 3.53 45.8 ± 27.00

M.G15.0.5 281.1 ± 9.79 42.8 ± 2.63

M.G15.3.10 215.6 ± 6.42 50.6 ± 4.45

C.G5.5.5 120.0 ± 54.45 27.9 ± 5.07

Diamicron® 211.7 ± 13.59

Page 87: Cassava starch as modified release excipient in selected

86

In table 4.4, the average MDT-values and similarity factor values of the different formulations

are given. This statistical analysis from the dissolution profile (figure 4.11 and table 4.4)

provided a more empirical analysis regarding the similarity between dissolution profiles of the

respective bead formulations and the control. It also provided data concerning the mean

dissolution time.

From table 4.4 it could be seen that, several of the formulations containing Avicel© had MDT

values notably longer (20 - 45%) than Diamicron®. A longer MDT indicates a slower rate of

release and a low MDT is indicative of a faster release. From the MDT values it could be seen

that M.G5.3.5 depicted a MDT 45.50% higher than Diamicron®. The cassava formulation

(C.G5.5.5) illustrated a dissolution profile, with an MDT of 43.60% less than that of the control.

This low MDT (119.99 ± 54.451) could be attributed to the rapid dissolution of the beads within

the first 15 min of the study. Formulation M.G15.3.10 exhibited a MDT-value similar to that of

Diamicron®. This correlates with the profile seen in figure 4.11; and the f2-value of M.G15.3.10

exceeded 50%, thus rendering M.G15.3.10 similar to the control. Although extended release for

formulation C.G5.5 can be observed from figure 4.11 it exhibited the fastest MDT value

(119.99 ± 54.541 min) and lowest similarity factor value (27.86 ± 5.071); and therefore it can be

concluded that the cassava-containing formula did exhibited a dissolution profile that differed

markedly from the profile observed for the control. Based on the dissolution parameters and

dissolution profiles depicted in figure 4.11, all formulations exhibited extended release although

to different degrees in comparison to the control, Diamicron® tablets. This highlights the

versatility of a multiple unit pellet system in modifying drug release.

HPMC has been described as an aid for modified drug release (Moodley et al., 2011:18-43;

Okunlola, 2015:1). This attribute could be ascribed to the high swellability and hydrophilic

nature of HPMC’s polymer matrix and consequent dissolution of the expanded matrix during

dissolution (Jiyauddin et al., 2014: Moodley et al., 2012:18-43; Oliveira et al., 2013:2; Scholtz

et al.¸ 2014:486-501; Siepmann & Peppas, 2012:163-173; Uhrich et al., 1999:3181-3198).

These various characteristics of HPMC contributed to prolonged MDT-values of the Avicel®

formulations.

4.7 SUMMARY

Moisture content as determined by Karl-Fischer titration indicated the starches were highly

susceptible to humidity. Conversely, powders with lower moisture content proved

Page 88: Cassava starch as modified release excipient in selected

87

advantageous in regards to flow. IR-spectroscopy indicated the relationship betweenthe two

starches and FTIR-analysis confirmed the difference between the two starch samples.

The bead formulations were evaluated with regards to surface morphology and internal

structure by means of SEM. SEM-micrographs, revealed that although not perfectly spherical,

beads were successfully prepared. The bead formulation containing cassava exhibited a rough

surface that could be accredited to the morphology of the cassava particles itself. Bead

formulations containing Avicel® demonstrated a more tightly packed internal structure. Size

analysis indicated that the majority of bead samples were larger than 1000 µm. Furthermore,

the starch powders exhibited poor powder flow properties, whereas all of the bead formulations

exhibited good powder flow properties. This was confirmed by all the powder flow parameters

that were determined.

With regards to swelling and erosion studies, the cassava-containing beads formulation

disintegrated quickly and swelling could therefore not be determined. However, Avicel® swelling

for all the Avicel®-containing formulations was evident. Moreover, it appeared that the inclusion

of HPMC resulted in an increased degree of bead swelling; however, its inclusion also resulted

in an increase in bead erosion.

Parameters and data obtained from the dissolution studies provided evidence which promotes

the application of cassava starch as an excipient in the production of modified release SODFs.

Depending on the excipients and manufacturing parameters used in production, the dissolution

behaviour can be similar to that of a commercial product or even be significantly greater.

Chapter 5 will conclude the study with a general summary and conclusion as well as possible

avenues for further study.

Page 89: Cassava starch as modified release excipient in selected

88

Chapter 5

GENERAL SUMMARY AND FUTURE PROSPECTS

5.1 SUMMARY & FUTURE PROSPECTS

The aim of this study as stated in Chapter 1, was to investigate the possible application of

cassava starch as a modified release excipient; and whether a modified SODF could be

manufactured. Chapter 2 provided a literature overview regarding SODF manufacturing,

different types of SODFs and excipients used to manufacture SODFs. It continued with the

description of starch as a versatile excipient and the selected starch source, cassava. Cassava,

one of the dominant sources of starch, is widely spread throughout sub-tropic environments and

easily cultivated. Being a biodegradable and renewable source of starch, cassava is promising

in many industries, e.g. clothing, paper, pharmaceutical, etc.. It is rich in amylopectin and

amylose, the two dominant biopolymers found within cassava. These polymers are cross-linked

forming a natural occurring matrix. Polymer-matrices have proven advantageous in the

development of various dosage forms, especially in modified release SODFs. For the above

mentioned reasons, the use of Cassava as a renewable source of starch can be advocated with

merit. Chapter 3 dealt with the experimental methods that were employed to determine the

physical characteristics, size, morphology and flow properties of the starch and extrusion-

spheronised beads.

From the results, it was evident that cassava starch was a hygroscopic starch, with weak flow

properties. This poor flowability would adversely affect the ability of the cassava starch to be

used in terms of direct compression. The moisture content of the powders was determined as

15.82 ± 0.339% and 11.84 ± 0.156% respectively for the donated and purchased starch. IR-

spectra were obtained to provide a fingerprint in order to compare the two starches. Thermo-

graphs were constructed for both starches and this indicated whether the moisture was present

as part of the chemical structure of the powder or whether it was present due to hygroscopicity.

From the thermo-graphs it could be seen that the moisture was present due to hygroscopicity

and not part of the chemical structure. This indicated the possibility of altering the powder flow

by simply heating the powder in a regulated oven.

By manufacturing beads from cassava the flow was drastically improved. The manufacturing of

cassava beads were difficult due to the presence of the hydrophobic drug, gliclazide, which was

Page 90: Cassava starch as modified release excipient in selected

89

selected as a model drug due to its importance as a second line treatment in Diabetes Mellitus

Type-2. Several successful bead formulations were manufactured with Avicel® as filler. The

production of Avicel® provided a product to which a bead manufactured frm cassava can be

compared. It was also evident from the study, that HPMC played a vital role in the manufacture

of quality beads. HPMC containing beads depicted a higher quality in terms of spherocity, which

corroborates findings by Dukiƈ-Ott et al. (2009:42-43) and Ghandi et al., (1999:166-168).

HPMC furthermore contributed to the modification of drug release (Moodley et al., 2012:21-36;

Oliveira et al., 2013:2). Multi-unit particulate system tablets could not be manufactured with

cassava beads due to the inability of the tablet to hold its shape. These tablets crumbled into

deformed beads. It was opted to encapsulate different intact bead samples in hard gelatine

capsules.

SEM-micrographs provided visual data relating to the shape of the powder particles, as well as

the whole beads, their internal structure and external morphology. These micrographs indicated

that the powder particles were spherical and exhibited surface irregularities. Size-distribution

analyses on the cassava powder particles and bead formulations were conducted. The D(0.9)

value for the starch particles were < 100 µm and all bead samples were < 1000 µm. Flow

properties of the powders and beads were characterised using various parameters (e.g. critical

orifice diameter, angle of repose, compressibility, etc.). Collectively, these parameters indicated

that the starches exhibited poor flow and that the beads portrayed acceptable flow. Friability

analyses illustrated that that the cassava bead formulations might not have acceptable physical

stability to withstand transport or handling.

Dissolution studies were conducted over a 12 h period. Samples were taken at predetermined,

time intervals and analysed with a UV-spectrometer for gliclazide content. Dissolution profiles

were characterised by means of the mean dissolution time (MDT) and similarity factor (f2).

Cassava starch beads provided modified drug release, where 60% of the total drug content was

pharmaceutically available within the first 15 min of the study. The remaining 40% dissolved

slowly over the remaining duration of the study. These beads depicted a MDT markedlyf less

than that of Diamicron®, at approximately 120.00 ± 13.59 min compared to a MDT value of

211.70 ± 13.59 min for the Diamicron® tablets. The release profile of the Cassava beads were

not similar to the control (Diamicron®) as evidenced by a similarity factor value of

27.86 ± 5.07%. Avicel® containing formulations exhibited MDT values ranging from

201.68 ± 5.34 to 307.05 ± 27.17 min. HPMC did not appear to affect drug release as no clear

tendency could be identified related to HPMC content.

Page 91: Cassava starch as modified release excipient in selected

90

5.2 FUTURE PROSPECTS

This study employed cassava starch, a sustainable, renewable and cost-effective, source of

starch as an excipient in modified release SODFs. Test formulations contained a binder, HPMC

and the poor water soluble drug, gliclazide. Extrusion-spheronisation was the chosen method of

bead manufacturing for the production of a modified release SODF. From the results of this

study, the following prospects for future studies are identified:

1) The effects of a water soluble drug can be investigated, instead of a poorly water soluble

drug, to evaluate the possible manufacturing of beads with cassava starch.

2) Multi-unit particulate system MUPS tablets containing cassava starch should be re-

manufactured, by altering the method of bead drying. Instead of using a regulated oven,

the beads could be lyophilised. This might provide a less rigid bead, capable of deforming

during compression and improving cohesion, which in turn could provide a more stable and

resilient MUPS tablet.

3) Another consideration in the manufacture of MUPS tablets from beads in this study, is the

mixing of a binder or filler to provide cohesion between the individual beads, before

compression.

4) Investigation of cassava starch in combination with other fillers, e.g. Avicel®, Microcelac®,

etc., in order to explore cassava starch as a release modifying excipient in bead

formulations and to evaluate the effect of these additions on bead manufacture.

5) An enteric coating can be applied to the cassava starch beads to prevent fast/immediate

dissolution of the cassava starch in the acidic environment of the stomach, which in turn

can influence the release profile of bead formulations containing this starch.

Page 92: Cassava starch as modified release excipient in selected

91

REFERENCES

I hereby confirm and declare that this study is my own and where

suitable reference and acknowledgement is given to external parties

and sources

~||~

ABDUL, S. & PODDAR, S. 2004. A flexible technology for modified release of drugs: multi

layered tablets. Journal of controlled release, 97:393-405.

ADDO, J., SMEETH, L. & LEON, D.A. 2007. Hypertension in Sub-Saharan Africa a systematic

review. Hypertension, 50:1012-1018.

AKHGARI, A., ABBASPOUR, M., REZAEE, S. & KUCHAK, A. 2007. Evaluation of the Swelling,

Erosion and Drug Release from Polysaccharide Matrix Tablets based on Pectin and Inulin.

Jundushapur journal of natural pharmaceutical products, 6:51-58.

ALDERBORN, G. 2002. Tablets and compaction. (In Aulton, M.E. ed. Pharmaceutics: the

science of dosage form design. 2nd ed. London: Churchill Livingstone. P. 397-440)

ALDERBORN, G. 2007. Tablets and compaction. (In Aulton, M.E. ed. Pharmaceutics: the

science of dosage form design. 3nd ed. London: Churchill Livingstone. P. 504-549)

ALLEN, L.V., Popovich, N.G., Ansel, H.C. 2011. Ansel’s Pharmaceutical dosage forms and drug

delivery systems. 9th ed. Philadelphia. Wolters Kluwer Health: Lippincott, Williams & Wilkins.

ASHFORD, M. 2007. Bioavailability – physicochemical and dosage form factors (In Aulton, M.E.

ed Pharmaceutics: the science of dosage form design. 3rd ed. Edinburgh: Churchill Livingstone).

AUCAMP, M., STIEGER, N., BARNARD, N. & LIEBENBERG, W. 2013. Solution-mediated

phase transformation of different roxithromycin solid-state forms: Implications on dissolution and

solubility. International journal of pharmaceutics, 449:18-27.

AULTON, M.E. & WELLS, T. 2002. Pharmaceutics: The science of dosage form design,

London: Churchill Livingstone.

Aulton, M.E., & Taylor, K.M. 2013. Aulton's pharmaceutics: the design and manufacture of

medicines. Elsevier Health Sciences

Page 93: Cassava starch as modified release excipient in selected

92

BADAWI, A.A. 2004. The Social Dimension of Globalization and Health. Perspectives on global

development and technology, 3:73-90.

BARDONNET, P., FAIVRE, V., PUGH, W., PIFFARETTI, J. & FALSON, F. 2006.

Gastroretentive dosage forms: Overview and special case of Helicobacter pylori. Journal of

controlled release, 111:1-18.

BARTELS, D. 2004. Adherence to oral therapy for type 2 diabetes: opportunities for enhancing

glycemic control. Journal of the american academy of nurse practitioners, 16:8-16

BAYOR, M. T., TUFFOUR, E. & LAMBON, P. S. 2013. Evaluation of Starch from New Sweet

Potato Genotypes for use as A Pharmaceutical Diluent, Binder or Disintegrant. Journal of

applied pharmaceutical science, 3:17-23

BENEKE, C. E., VILJOEN, A. M. & HAMMAN, J. H. 2009. Polymeric plant-derived excipients in

drug delivery. Molecules, 14:2602-2620.

BHASKARAN, S. & LAKSHMI, P. 2010. Extrusion spheronization—A review. International

journal of pharmeutical technology research,2:2429-2433.

BRITISH PHARMACOPOEIA. 2015. https://www.pharmacopoeia.com/BP2015

Date of access: 30 October 2015

BUDIASIH, S., JIYAUDDIN, K., LOGAVINOD, N., KALEEMULLAH, M., JAWAD, A., SAMER,

A., FADLI, A. & EDDY, Y. Optimization of Polymer Concentration for Designing of Oral Matrix

Controlled Release Dosage Form. UK journal of pharmaceutical and biosciences, 2:54-61

BÜHLER, V. 2003. Kollidon® Polyvinylpyrrolidone for the pharmaceutical industry 7th ed.

Ludwigshaven: BASF

BUYS, G.M. 2005. Formulation of a chitosan multi-dosage form for drug delivery to the colon.

Potchefstroom North-West University (Thesis-PhD).

CASAS, M., FERRERO, C. & JIMÉNEZ-CASTELLANOS, M.R. 2010. Graft tapioca starch

copolymers as novel excipients for controlled-release matrix tablets. CarbohydratepPolymers,

80:71-77.

CASSAVA IMAGE https://theglyptodon.files.wordpress.com/2011/06/cassava.jpg Date of

access: 30 March 2014

Page 94: Cassava starch as modified release excipient in selected

93

CHARLES, A.L., CHANG, Y.H., KO, W.C., SRIROTH, K. & HUANG, T.C. 2005. Influence of

amylopectin structure and amylose content on the gelling properties of five cultivars of cassava

starches. Journal of agricultural and food chemistry, 53:2717-2725.

CHEBOYINA, S. & O'HAVER, J.H. 2004. Process and apparatus for producing spherical pellets

using molten solid matrices. Google Patents.

CHINYEMBA, P. 2012. Use of Aloe vera and Aloe marlothii materials as excipients in beads

produced by extrusion-spheronization. Potchefstroom North-West University (Dissertation –

MSc).

CHITEDZE, J., MONJEREZI, M., SAKA, J. & STEENKAMP, J. 2012. Binding Effect of Cassava

Starches on the Compression and Mechanical Properties of Ibuprofen Tablets. Journal of

applied pharmaceutical sciences, 4:31-37

COLOMBO, P., BETTINI, R., SANTI, P. & PEPPAS, N.A. 2000. Swellable matrices for

controlled drug delivery: gel-layer behaviour, mechanisms and optimal performance.

Pharmaceutical science and technology today, 3:198-204.

CHOPRA, S., VENKATESAN, N. & BETAGRI, G.V. 2013 Formulation of lipid bearing pellets

as a delivery system for poorly soluble drugs. International journal of pharmaceutics,

446:136-144

CORDAIN, L., EATON, S.B., SEBASTIAN, A., MANN, N., LINDEBERG, S., WATKINS, B.A.,

O’KEEFE, J.H. & BRAND-MILLER, J. 2005. Origins and evolution of the Western diet: health

implications for the 21st century. The American journal of clinical nutrition, 81:341-354.

COSTA, P. & LOBO, J. M. S. 2001. Modeling and comparison of dissolution profiles. European

journal of pharmaceutical sciences, 13:123-133.

CROUTER, A. & BRIENS, L. 2014. The effect of moisture on the flowability of pharmaceutical

excipients. American association of pharmaceutical scientist PharmSciTech, 15:65-74.

DALL, T.M., ZHANG, Y., CHEN, Y.J., QUICK, W.W., YANG, W.G. & FOGLI, J. 2010. The

economic burden of diabetes. Health affairs, 29:297-303.

DAS, N.G. & DAS, S.K. 2003. Controlled release of oral dosage forms. Pharmaceutical

technology, 15:10-17.

Page 95: Cassava starch as modified release excipient in selected

94

DAY, C. & BAILEY, C.J. 2011. Obesity in the pathogenesis of type 2 diabetes. The british

journal of diabetes and vascular disease, 11:55-61.

DE KOCK, J.M. 2005. Chitosan as a multipurpose excipient in directly compressed minitablets.

North-West University (Thesis – PhD).

DE FLOOR, I., DEHING, I. & DELCOUR, J. 1998. Physico‐Chemical Properties of Cassava

Starch. Starch‐Stärke, 50:58-64.

DELAMATER, A.M. 2006. Improving patient adherence. Clinical diabetes, 24:71-77.

DEY, N.C., MAJUMDAR, S. & RAO, M.E.B. 2008 Multiparticulate Drug Delivery Systems for

Controlled Release. Tropical journal of pharmaceutical research, 7:1067-1075

DHANDAPANI, N.V. 2012. Pelletization by Extrusion-Spheronization-A detailed review. The all

results journals: Biology, 3:10-23.

DIABETES http://www.who.int/diabetes/facts/en/ Date of access: 25 August 2013

DRESSER, G.K., SPENCE, J.D. & BAILEY, D.G. 2000. Pharmacokinetic-pharmacodynamic

consequences and clinical relevance of cytochrome P450 3A4 inhibition. Clinical

pharmacokinetics, 38:41-57.

DRESSMAN, J.B., AMIDON, G L., REPPAS, C. & SHAH, V.P. 1998. Dissolution testing as a

prognostic tool for oral drug absorption: immediate release dosage forms. Pharmaceutical

research, 15:11-22.

DUKIĆ-OTT, A., THOMMES, M., REMON, J.P., KLEINEBUDDE, P. & VERVAET, C. 2009.

Production of pellets via extrusion–spheronisation without the incorporation of microcrystalline

cellulose: a critical review. European journal of pharmaceutics and biopharmaceutics, 71:38-46.

DUMOULIN, Y., CARRIERE, F. & INGENITO, A. 1998. Manufacture of cross-linked amylose

useful as an excipient for control release of active compounds. Google Patents.

DURAIAPPAH, A. K. 1998. Poverty and environmental degradation: a review and analysis of

the nexus. World development, 26:2169-2179.

EMERY, E., OLIVER, J., PUGSLEY, T., SHARMA, J. & ZHOU, J. 2009. Flowability of moist

pharmaceutical powders. Powder technology, 189:409-415.

Page 96: Cassava starch as modified release excipient in selected

95

FALDU, B. & ZALAVADIYA, B. 2012. LUBRICANTS: FUNDAMENTALS OF TABLET

MANUFACTURING. International journal of research in pharmacy and chemistry, 2:2231-

2781.FAMÁ, L., FLORES, S.K., GERSCHENSON, L. & GOYANES, S. 2006. Physical

characterization of cassava starch biofilms with special reference to dynamic mechanical

properties at low temperatures. Carbohydrate polymers, 66:8-15.

FAMÁ, L., GOYANES, S. & GERSCHENSON, L. 2007. Influence of storage time at room

temperature on the physicochemical properties of cassava starch films. Carbohydrate olymers,

70:265-273.

FOWLER, M.J. 2007. Diabetes treatment, part 2: oral agents for glycemic management. Clinical

diabetes, 25:131-134.

FRIZON, F., DE OLIVEIRA ELOY, J., DONADUZZI, C.M., MITSUI, M.L. & MARCHETTI, J.M.

2013. Dissolution rate enhancement of loratadine in polyvinylpyrrolidone K-30 solid dispersions

by solvent methods. Powder technology, 235:532-539.

Gandhi, B., & Baheti, J. 2013. Multiparticulates drug delivery systems: a review. Indian journal

of pharmaceutical chemical sciences, 2:1620-6.

GANDHI, R., KAUL, C. L. & PANCHAGNULA, R. 1999. Extrusion and spheronization in the

development of oral controlled-release dosage forms. Pharmaceutical science and technology

today, 2:160-170.

GARR, J. & RUBINSTEIN, M. 1992. The influence of moisture content on the consolidation and

compaction properties of paracetamol. International journal of pharmaceutics, 81:187-192.

GELDART, D., ABDULLAH, E., HASSANPOUR, A., NWOKE, L. & WOUTERS, I. 2006.

Characterization of powder flowability using measurement of angle of repose. China

particuology, 4:104-107.

GHORI, M. U., GINTING, G., SMITH, A.M. & CONWAY, B.R. 2014. Simultaneous quantification

of drug release and erosion from hypromellose hydrophilic matrices. International journal of

pharmaceutics, 465:405-412.

GOYAL, A., SHUKLA, P. & SRIVASTAVA, A. 2009. Factors influencing drug release

characteristic from hydrophilic polymer matrix tablet. Asian journal of pharmcological clinic

research, 2:93-98.

Page 97: Cassava starch as modified release excipient in selected

96

GUPTA, B.P., THAKUR, N., JAIN, N.P., BANWEER, J. & JAIN, S. 2010. Osmotically controlled

drug delivery system with associated drugs. Journal of pharmacy and pharmaceutical sciences,

13:571-588.

HALL, V., THOMSEN, R.W., HENRIKSEN, O. & LOHSE, N. 2011. Diabetes in Sub Saharan

Africa 1999-2011: epidemiology and public health implications. A systematic review. BMC public

health, 11:564.

HANCOCK, B.C., VUKOVINSKY, K.E., BROLLEY, B., GRIMSEY, I., HEDDEN, D., OLSOFSKY,

A. & DOHERTY, R. A. 2004. Development of a robust procedure for assessing powder flow

using a commercial avalanche testing instrument. Journal of pharmaceutical and biomedical

analysis, 35:979-990.

HART, A. 2015. Effect of Particle Size on Detergent Powders Flowability and Tabletability.

Journal of chemical engineering and process technology, 6:1.

HIRANI, J.J., RATHOD, D.A. & VADALIA, K.R. 2009. Orally disintegrating tablets: a review.

Tropical journal of pharmaceutical research, 8.

HORN, E. 2008. Development of a composite index for pharmaceutical powders. Potchefstroom

North-West University (Dissertation-MSc)

HUANG, X. & BRAZEL, C.S. 2001. On the importance and mechanisms of burst release in

matrix-controlled drug delivery systems. Journal of controlled release, 73:121-136.

HUANG, Z.Q., LU, J.P., LI, X.H. & TONG, Z.F. 2007. Effect of mechanical activation on physico-

chemical properties and structure of cassava starch. Carbohydrate Polymers, 68:128-135.

IBRAGIMOVA, M. M. & IKRAMOV, L.T. 2015. Analysis of Glimepiride in Human Blood and

Urine by Thin-Layer Chromatography and UV-Spectrophotometry. The bulletin of legal

medicine, 20:71-75.

INGLE, L.M., WANKHADE, V.P., UDASI, T.A. & TAPAR, K.K. 2013. New Approaches for

Development and Characterization of SMEDDS. International journal of pharmacy and

pharmaceutical science research, 3:7-14.

ISPAS-SZABO, P., RAVENELLE, F., HASSAN, I., PREDA, M. & MATEESCU, M.A. 1999.

Structure–properties relationship in cross-linked high-amylose starch for use in controlled drug

release. Carbohydrate research, 323:163-175.

Page 98: Cassava starch as modified release excipient in selected

97

JALLO, L.J., GHOROI, C., GURUMURTHY, L., PATEL, U. & DAVÉ, R.N. 2012. Improvement of

flow and bulk density of pharmaceutical powders using surface modification. International

journal of pharmaceutics, 423:213-225.

JAMADAR, S.A., MULYE, S.P., KAREKAR, P.S., PORE, Y.V. & BURADE, K.B. 2011. Development and

validation of UV spectrophotometric method for the determination of Gliclazide in tablet dosage form.

Der pharma chemical, 3:338-343

JAVADZADEH, Y., VAZIFEHASL, Z., DIZAJ, S.M. & MOKHTARPOUR, M. 2015. Spherical

Crystallization of Drugs. Advanced topics in crystallization, Chapter 4:85-104.

JEWESSON, P.J. 1996. Economic impact of intravenous-to-oral antibacterial stepdown therapy.

Clinical drug investigation, 11:1-9.

JIVRAJ, M., MARTINI, L.G. & THOMSON, C. M. 2000. An overview of the different excipients

useful for the direct compression of tablets. Pharmaceutical science andtechnology today, 3:58-

63.

JIYAUDDIN, K., SUNG, K., SAMER, A., KALEEMULLAH, M., RASHA, S., BUDIASIH, S.,

JAWAD, A., RASNY, R., GAMAL, E. & JUNAINAH, H. 2014. Comparative study on the effect of

hydrophilic and hydrophobic polymers on the dissolution rate of a poorly water soluble drug. UK

journal of pharmaceutical and biosciences, 2:51-61

JOSHI, A.K., PUND, S.V., NIVSARKAR, M.A., VASU, K.K. & SHISHOO, C.J. 2011. Exploring

the potential of polacrilin potassium as a novel super disintegrant in microcrystalline cellulose

based pellets prepared by extrusion-spheronization. Chronicles of young scientists, 2:111.

KALRA, S. & GUPTA, Y. 2015. Sulfonylureas. The journal of the pakistan medical association,

65:101-104.

KALRA, S., MADHU, S. & BAJAJ, S. 2015. Sulfonylureas: Assets in the past, present and

future. Indian journal of endocrinology and metabolism, 19:314.

KARDAS, P. 2005. The DIACOM study (effect of Dosing frequency of oral Antidiabetic agents

on the Compliance and biochemical control of type 2 diabetes). Diabetes, Obesity and

Metabolism, 7:722-728.

Page 99: Cassava starch as modified release excipient in selected

98

KASSAB, N.M., AMARAL, M.S.D., SINGH, A.K. & SANTORO, M.I.R.M. 2010. Development and

validation of UV spectrophotometric method for determination of levofloxacin in pharmaceutical

dosage forms. Química Nova, 33:968-971.

KATZUNG, B., MASTERS, S. & TREVOR, A. 2009. Basic and Clinical Pharmacology. McGraw-

Hill Companies. Inc., New York.

KAWACHI, I., KENNEDY, B.P., LOCHNER, K. & PROTHROW-STITH, D. 1997. Social capital,

income inequality, and mortality. American journal of public health, 87:1491-1498.

KHAN, A., MALVIYA, R. & SHARMA, P.K. 2014. Multi Unit Drug Delivery System–A Brief

Review of Pelletization Technique. World applied sciences journal, 31:2137-2140.

KHAN, G.M. 2001. Controlled release oral dosage forms: Some recent advances in matrix type

drug delivery systems. The sciences, 1:350-354.

KIM, E. H.J., CHEN, X.D. & PEARCE, D. 2005. Effect of surface composition on the flowability

of industrial spray-dried dairy powders. Colloids and surfaces b: biointerfaces, 46:182-187.

KO, K.H., RAWAL, A. & SAHAJWALLA, V. 2014. Analysis of thermal degradation kinetics and

carbon structure changes of co-pyrolysis between macadamia nut shell and PET using

thermogravimetric analysis and 13 C solid state nuclear magnetic resonance. Energy

conversion and management, 86:154-164.

KOESTER, M. AND THOMMES, M. 2010. New insights into the pelletization mechanism by extrusion/epheronization. American association of pharmaceutical scientist PharmSciTech, 11:1549-1551.

KORHONEN, O., RAATIKAINEN, P., HARJUNEN, P., NAKARI, J., SUIHKO, E., PELTONEN,

S., VIDGREN, M. & PARONEN, P. 2000. Starch acetates—multifunctional direct compression

excipients. Pharmaceutical research, 17:1138-1143.

KRAJACIC, A. & TUCKER, I.G. 2003. Matrix formation in sustained release tablets: possible

mechanism of dose dumping. International journal of pharmaceutics, 251:67-78.

KRULL, E.S., MCBEATH, A.V., SMERNIK, R.J. AND LEHMANN, J. 2014. The influence of feedstock and production temperature on biochar carbon chemistry: A solid-state 13C NMR study. Biomass and bioenergy, 60:121-129

Page 100: Cassava starch as modified release excipient in selected

99

KUMAR, S., DAS, B. & RAJU, S.R. 2012. Formulation and Evaluation of Multiunit Pellet System

of Venlafaxine Hydrochloride. Journal of pharmaceutical and biomedical sciences, 18:182-3.

LAHIRI, S.W. 2012. Management of type 2 diabetes: what is the next step after metformin?

Clinical diabetes, 30:72-75.

LAJOINIE, A., HENIN, E., KASSAI, B. & TERRY, D. 2014. Solid oral forms availability in

children: A cost saving investigation. British journal of clinical pharmacology, 78:1080-1089.

LAMBRECHTS, J.J. 2008. Investigation into the influence of different Kollidon polymers on the

properties of powder mixtures intended for tableting. Potchefstroom North-West University

(Dissertation – MSc).

LANDILLON, V., CASSAN, D., MOREL, M.H. & CUQ, B. 2008. Flowability, cohesive, and

granulation properties of wheat powders. Journal of food engineering, 86:178-193.

LAVANYA, K., SENTHIL, V. & RATHI, V. 2011. Pelletization technology: a quick review.

International journal of pharmaceutical scientific research, 2:1337-1355.

LAVOIE, F., CARTILIER, L. & THIBERT, R. 2002. New methods characterizing avalanche

behavior to determine powder flow. Pharmaceutical research, 19:887-893.

LAZAR, M. A. 2005. How obesity causes diabetes: not a tall tale. Science, 307:373-375.

LEBOVITZ, H. E. 1999. Type 2 diabetes: an overview. Clinical chemistry, 45:1339-1345.

LEMIEUX, M., GOSSELIN, P. & MATEESCU, M.A. 2009. Carboxymethyl high amylose starch

as excipient for controlled drug release: Mechanistic study and the influence of degree of

substitution. International journal of pharmaceutics, 382:172-182.

LEMMER, H., STIEGER, N., LIEBENBERG, W. & CAIRA, M.R. 2012. Solvatomorphism of the

antibacterial dapsone: x-ray structures and thermal desolvation kinetics. Crystal growth &

design, 12:1683-1692.

LENAERTS, V., DUMOULIN, Y. & MATEESCU, M.A. 1991. Controlled release of theophylline

from cross-linked amylose tablets. Journal of controlled release, 15:39-46.

LENAERTS, V., MOUSSA, I., DUMOULIN, Y., MEBSOUT, F., CHOUINARD, F., SZABO, P.,

MATEESCU, M., CARTILIER, L. & MARCHESSAULT, R. 1998. Cross-linked high amylose

Page 101: Cassava starch as modified release excipient in selected

100

starch for controlled release of drugs: recent advances. Journal of controlled release, 53:225-

234.

LIU, L. X., MARZIANO, I., BENTHAM, A., LITSTER, J.D., WHITE, E. & HOWES, T. 2008. Effect

of particle properties on the flowability of ibuprofen powders. International journal of

pharmaceutics, 362:109-117.

LOURENÇO, F.R., GHISLENI, D.D.M., YAMAMOTO, R.N. & PINTO, T.D.J.A. 2013.

Comparison of dissolution profile of extended-release oral dosage forms-two one-sided

equivalence test. Brazilian journal of pharmaceutical sciences, 49:367-371.

MALLIPEDDI, R., SARIPELLA, K.K. & NEAU, S.H. 2010. Use of coarse ethylcellulose and PEO

in beads produced by extrusion–spheronization. International journal of pharmaceutics, 385:53-

65.

MALLIPEDDI, R., SARIPELLA, K. K. & NEAU, S. H. 2014. Use of fine particle ethylcellulose as

the diluent in the production of pellets by extrusion-spheronization. Saudi pharmaceutical

journal, 22:360-372.

MANDAL, S., BASU, S.K. & SA, B. 2009. Sustained release of a water-soluble drug from

alginate matrix tablets prepared by wet granulation method. American association of

pharmaceutical scientists PharmSciTech, 10:1348-1356.

MARAIS, E.B. 2013 Permeation of excised intestinal tissue by insulin released from Eudragit®

L100/Trimethyl chitosan chloride microspheres. Potchefstroom North-West University

(Dissertation-MSc).

Martin AN. Physical Pharmacy: Physical Chemical Principles in the Pharmaceutical Sciences,

4th edition, Lippincott Williams & Wilkins: USA, 1993.

MCCULLOCH, D.K., MUNSHI, M., NATHAN, D. M., SCHMADER, K. E. & MULDER, J. E.

Treatment of type 2 diabetes mellitus in the older patient.

MEYROWITSCH, D.W. & BYGBJERG, C. 2007. Global burden of disease-a race against time.

Danish medical bulletin, 54:32-4.

MILLILI, G. & SCHWARTZ, J. 1990. The strength of microcrystalline cellulose pellets: the effect

of granulating with water/ethanol mixtures. Drug development and industrial pharmacy,

16:1411-1426.

Page 102: Cassava starch as modified release excipient in selected

101

Molecular and macroscopic structure of amylose and amylopectin.

http://voer.edu.vn/m/carbohydrates/70e79f40 Date of access: 30 March 2014

MOODLEY, K., PILLAY, V., CHOONARA, Y. E., DU TOIT, L C., NDESENDO, V.M., KUMAR,

P., COOPPAN, S. & BAWA, P. 2011. Oral drug delivery systems comprising altered geometric

configurations for controlled drug delivery. International journal of molecular sciences, 13:18-43.

MOORE, J.W. & FLANNER, H.H. 1996. Mathematical comparison of dissolution profiles.

Pharmaceutical technology, 20, 64-74.

MOORTHY, S.N. 2002. Physicochemical and functional properties of tropical tuber starches: a

review. Starch‐Stärke, 54:559-592.

MOTALA, A. & RAMAIYA, K. 2010. Diabetes: the hidden pandemic and its impact on Sub-

saharan Africa. Diabetes leadership forum

MULLARNEY, M.P., BEACH, L.E., DAVÉ, R.N., LANGDON, B.A., POLIZZI, M. &

BLACKWOOD, D.O. 2011. Applying dry powder coatings to pharmaceutical powders using a

co-mill for improving powder flow and bulk density. Powder tTechnology, 212:397-402.

MULTIPARTICULATE DRUG DELIVERY SYSTEMS.

http://www.slideshare.net/bknanjwade/multiparticulate-drug-delivery-systems Date of access:

25 April 2014

MUSTAFA, M.E., NUR, A.O., OSMAN, Z.A. & AHMED, S.A. 2014. Influence of drug solubility

and polymers supply source on the physical performance of matrix tablets. International journal

of pharmacy and pharmaceutical sciences, 6:301-312

NEWTON, J., CHAPMAN, S. & ROWE, R. 1995. The influence of process variables on the

preparation and properties of spherical granules by the process of extrusion and spheronisation.

International journal of pharmaceutics, 120:101-109.

NOKHODCHI, A. 2005. Effect of moisture on compaction and compression. Pharmaceutical

Technology, 646-66.

NUWAMANYA, E., BAGUMA, Y., EMMAMBUX, N., TAYLOR, J. & PATRICK, R. 2010.

Physicochemical and functional characteristics of cassava starch in Ugandan varieties and their

progenies. Journal of plant breeding and crop science, 2:001-011.

Page 103: Cassava starch as modified release excipient in selected

102

OKUNLOLA, A., ODEKU, O.A. & PATEL, R. P. 2012. Formulation optimization of floating

microbeads containing modified Chinese yam starch using factorial design. Journal of excipients

and food chemicals, 3:17-25.

OLIVEIRA, P.R., MENDES, C., KLEIN, L., SANGOI, M.D.S., BERNARDI, L. S. & SILVA, M.A.

S. 2013. Formulation development and stability studies of norfloxacin extended-release matrix

tablets. BioMed research international, 2013:1-9

Oxford dictionary definition of morphology.

http://www.oxforddictionaries.com/definition/english/morphology Date of access: 20 March

2014.

PAINE, M F., HART, H.L., LUDINGTON, S.S., HAINING, R.L., RETTIE, A.E. & ZELDIN, D.C.

2006. The human intestinal cytochrome P450 “pie”. Drug metabolism and disposition, 34:880-

886.

PANTEN, U., SCHWANSTECHER, M. AND SCHWANSTECHER, C. 1996. Sulfonylurea

receptors and mechanism of sulfonylurea action. Experimental and clinical endocrinology

and diabetes, 104:1-9

PATEL, A., RAY, S. & THAKUR, R.S. 2006. Invitro evaluation and optimization of controlled

release floating drug delivery system of metformin hydrochloride. DARU Journal of

pharmaceutical sciences, 14:57-64.

PATEL, P.S., RAVAL, J. P. & PATEL, H.V. 2010. Review on the pharmaceutical applications of

hot melt extruder. Asian journal of pharmaceutical and clinical research, 3.

PERIOLI, L., D’ALBA, G. & PAGANO, C. 2012. New oral solid dosage form for furosemide oral

administration. European journal of pharmaceutics and biopharmaceutics, 80:621-629.

Physicochemical properties of gliclazide.

http://www.chemicalbook.com/ChemicalProductProperty_EN_CB5113462.htm Date of access:

30 May 2014

Physicochemical properties of gliclazide. http://www.drugbank.ca/drugs/DB01120 Date of

access: 30 May 2014

Radial extruder. http://spheronizer.com/html/extrusion.html Date of access: 20 April 2014

Page 104: Cassava starch as modified release excipient in selected

103

REDDY, S.G. 2006. Globalisation, labour markets, and social outcomes in developing countries.

Available at SSRN 942087.

Reddy, R.B., Malleswari, K., Prasad, G., & Pavani, G. 2012. Colon targeted drug delivery

system: a review. International journal of pharmaceutical science and research, 4:42-54.

REMKO, M. 2009. Theoretical study of molecular structure, pKa, lipophilicity, solubility,

absorption, and polar surface area of some hypoglycemic agents. Journal of molecularstructure:

THEOCHEM, 897:73-82.

RENDELL, M. 2004. The role of sulphonylureas in the management of type 2 diabetes mellitus.

Drugs, 64:1339-1358.

REPPAS, C. & NICOLAIDES, E. 2000. Analysis of drug dissolution data. Drugs and the

pharmaceutical sciences, 106:229-254.

ROLLAND-SABATÉ, A., SÁNCHEZ, T., BULÉON, A., COLONNA, P., JAILLAIS, B.,

CEBALLOS, H. & DUFOUR, D. 2012. Structural characterization of novel cassava starches with

low and high-amylose contents in comparison with other commercial sources. Food

hydrocolloids, 27:161-174.

ROY, S., RIGA, A. & ALEXANDER, K. 2002. Experimental design aids the development of a

differential scanning calorimetry standard test procedure for pharmaceuticals. Thermochimica

acta, 392:399-404.

SAHOO, S., PARVEEN, S. & PANDA, J. 2007. The present and future of nanotechnology in

human health care. Nanomedicine: Nanotechnology, Biology and medicine, 3:20-31.

SASTRY, S.V., NYSHADHAM, J.R. & FIX, J.A. 2000. Recent technological advances in oral

drug delivery–a review. Pharmaceutical science and technology today, 3:138-145.

Save and Grow Cassava: A guide to sustainable production 2013.

http://www.fao.org/docrep/018/i3278e/i3278e.pdf Date of access: 30 August 2013

SCHIELE, J.T., QUINZLER, R., KLIMM, H.D., PRUSZYDLO, M.G. & HAEFELI, W.E. 2013.

Difficulties swallowing solid oral dosage forms in a general practice population: prevalence,

causes, and relationship to dosage forms. European journal of clinical pharmacology, 69:937-

948.

Page 105: Cassava starch as modified release excipient in selected

104

SCHOLTZ, J., VAN DER COLFF, J., STEENEKAMP, J., STIEGER, N. & HAMMAN, J. 2014.

More good news about polymeric plant-and algae-derived biomaterials in drug delivery systems.

Current drug targets, 15:486-501.

SHAH, A. & MLODOZENIEC, A. 1977. Mechanism of surface lubrication: Influence of duration

of lubricant‐excipient mixing on processing characteristics of powders and properties of

compressed tablets. Journal of pharmaceutical sciences, 66:1377-1382.

SIEGEL, R.A. & RATHBONE, M. J. 2012. Overview of controlled release mechanisms.

Fundamentals and applications of controlled release drug delivery. Springer.

SIEPMANN, J. & PEPPAS, N. 2012. Modeling of drug release from delivery systems based on

hydroxypropyl methylcellulose (HPMC). Advanced drug delivery reviews, 64:163-174.

SIEWERT, M., DRESSMAN, J., BROWN, C. K., SHAH, V.P., AIACHE, J.M., AOYAGI, N.,

BASHAW, D., BROWN, C., BROWN, W. & BURGESS, D. 2003. FIP/AAPS guidelines to

dissolution/in vitro release testing of novel/special dosage forms. American association of

pharmaceutical scientists PharmSciTech, 4:43-52.

SINGH, K., KUMAR, A., LANGYAN, N. & AHUJA, M. 2009. Evaluation of Mimosa pudica seed

mucilage as sustained-release excipient. American association of pharmaceutical scientists

PharmSciTech, 10:1121-1127.

SONNEKUS, J. 2008. Characterization of the flow and compression properties of chitosan.

Potchefstroom North-West University (Dissertation-MSc).

Social determinants of health 2007

http://www.wpro.who.int/health_research/documents/dhs_hr_health_in_asia_and_the_pacific_0

7_chapter_2_social_determinants_of_health.pdf?ua=1 Date of access: 20 August 2013

STANIFORTH, J.N. 2002. Powders comprising anti-adherant materials for use in Dry Powder

Inhalers. Google Patents.

STANIFORTH, J.N. & AULTON, M.E. 2007. Particle size analysis (In Aulton, M.E. ed

Pharmaceutics: the science of dosage form design. 3rd ed. Edinburgh: Churchill Livingstone).

STORPER, M. 2000. Lived effects of the contemporary economy: globalization, inequality, and

consumer society. Public culture:12, 375-409.

Page 106: Cassava starch as modified release excipient in selected

105

SUMMERS, M.P & AULTON, M.E. 2007. Granulation (In Aulton, M.E. ed Pharmaceutics: the

science of dosage form design. 3rd ed. Edinburgh: Churchill Livingstone).

Sungthongieen, S., Sriamornsak, P., Pitaksuteepong, T., Somsiri, A., & Puttipipatkhachorn, S.

2004. Effect of degree of esterification of pectin and calcium amount on drug release from

pectin-based matrix tablets. American association of pharmaceutical scientist PharmSciTech, 5,

50–57.

TAKSANDE, A., TAKSANDE, B., KUMAR, A. & VILHEKAR, K. 2008. Malnutrition related

diabetes mellitus. The journal of Mahatma Gandhi institute. 13:19-24

TEUNOU, E. & FITZPATRICK, J. 1999. Effect of relative humidity and temperature on food

powder flowability. Journal of food engineering, 42:109-116.

The great exchange. 2008. http://www.vos.noaa.gov/MWL/dec_08/great_exchange.shtml Date

of access: 30 August 2013

TONUKARI, N.J. 2004. Cassava and the future of starch. Electronic journal of biotechnology,

7:5-8.

TOUSEY, M.D. 2002. The granulation process 101. Pharmaceutical technology, 8-13.

Traina, K., CLOOTS, R., BONTEMPI, S., LUMAY, G., VANDEWALLE, N. AND BOSCHINI, F.

2013. Flow abilities of powders and granular materials evidenced from dynamical tap density

measurement. Powder technology, 235:842-852.

UHRICH, K.E., CANNIZZARO, S.M., LANGER, R.S. & SHAKESHEFF, K.M. 1999. Polymeric

systems for controlled drug release. Chemical reviews, 99:3181-3198.

United States Pharmacopoiea. www.usp.org Date of access 2014 & 2015

VANDERPOEL, D.R., HUSSEIN, M.A., WATSON-HEIDARI, T. & PERRY, A. 2004. Adherence

to a fixed-dose combination of rosiglitazone maleate/metformin hydrochloride in subjects with

type 2 diabetes mellitus: a retrospective database analysis. Clinical therapeutics:26, 2066-2075.

VELASCO, M., MUNOZ-RUIZ, A., MONEDERO, M. & JIMÉNEZ-CASTELLANOS, M. 1995.

Study of flowability of powders. effect of the addition of lubricants. Drug development and

industrial pharmacy, 21:2385-2391.

Page 107: Cassava starch as modified release excipient in selected

106

VERVAET, C., BAERT, L. & REMON, J. P. 1995. Extrusion-spheronisation A literature review.

International journal of pharmaceutics, 116:131-146.

VICENTINI, N., DUPUY, N., LEITZELMAN, M., CEREDA, M. & SOBRAL, P. 2005. Prediction of

cassava starch edible film properties by chemometric analysis of infrared spectra. Spectroscopy

letters, 38:749-767.

VILJOEN, J.M., STEENEKAMP, J.H., MARAIS, A.F. & KOTZÉ, A.F. 2014. Effect of moisture

content, temperature and exposure time on the physical stability of chitosan powder and tablets.

Drug development and industrial pharmacy, 40:730-742.

VIRIDÉN, A., ABRAHMSÉN-ALAMI, S., WITTGREN, B. & LARSSON, A. 2011. Release of

theophylline and carbamazepine from matrix tablets–consequences of HPMC chemical

heterogeneity. European journal of pharmaceutics and biopharmaceutics, 78:470-479.

VIRIDÉN, A., LARSSON, A., SCHAGERLÖF, H. & WITTGREN, B. 2010. Model drug release

from matrix tablets composed of HPMC with different substituent heterogeneity. International

journal of pharmaceutics, 401:60-67.

WAGSTAFF, A. 2002. Poverty and health sector inequalities. Bulletin of the world health

organization, 80:97-105.Diabetes fact sheet 2013.

http://www.who.int/diabetes/facts/world_figures/en Date of access: 3 March 2013

WILSON, I.D., NICHOLSON, J.K., CASTRO-PEREZ, J., GRANGER, J.H., JOHNSON, K.A., SMITH, B.W. & PLUMB, R.S. 2005. High resolution “ultra performance” liquid chromatography coupled to oa-TOF mass spectrometry as a tool for differential metabolic pathway profiling in functional genomic studies. Journal of proteome research, 4:591-599

WONG, A. C.-Y. 2002. Use of angle of repose and bulk densities for powder characterization

and the prediction of minimum fluidization and minimum bubbling velocities. Chemical

engineering science, 57:2635-2640.

YORK, P. 2013. Design of dosage forms. Aulton's pharmaceutics: The design and manufacture

of medicines, 7.

YOUNG, C.R., KOLENG, J.J. & MCGINITY, J.W. 2002. Production of spherical pellets by a hot-

melt extrusion and spheronization process. International journal of pharmaceutics, 242:87-92.

Page 108: Cassava starch as modified release excipient in selected

107

YANG, M., XIE, S., LI, Q., WANG, Y., CHANG, X., SHAN, L., SUN, L., HUANG, X. & GAO, C.,

2012. Effects of polyvinylpyrrolidone both as a binder and pore-former on the release of

sparingly water-soluble topiramate from ethylcellulose coated pellets. International journal of

pharmaceutics, 465:187-196

ZHANG, G.G., LAW, D., SCHMITT, E.A. & QIU, Y. 2004. Phase transformation considerations

during process development and manufacture of solid oral dosage forms. Advanced drug

delivery reviews, 56:371-390.

ZHANG, Y., LAW, Y. & CHAKRABARTI, S. 2003. Physical properties and compact analysis of

commonly used direct compression binders. aaps Pharmscitech, 4:489-499.

ZIMMET, P., ALBERTI, K. & SHAW, J. 2001. Global and societal implications of the diabetes

epidemic. Nature, 414:782-787.

Page 109: Cassava starch as modified release excipient in selected

108

ANNEXURE A

THERMOANALYSIS, MOISTURE CONTENT AND SIZE-DISTRIBUTION

Page 110: Cassava starch as modified release excipient in selected

109

MOISTURE CONTENT

Table A.I: Karl-Fischer titration values for moisture content of the donated Cassava starch

Donated Starch

Time

(min)

Moisture (%H2O) %Weight

Loss Run 1 Run 2 Average SD %RSD

Reference Sample

0 15.58 16.06 15.82 0.339 0.24 10.80

Moisture Content at 25°C

30 14.15 14.11 14.13 0.028 0.02 10.21

60 12.89 12.88 12.89 0.007 0.01 5.75

120 11.94 12.1 12.02 0.113 0.08 5.63

240 11.61 11.65 11.63 0.028 0.02 9.167

360 11.37 11.71 11.54 0.240 0.17 8.84

480 11.49 11.98 11.74 0.346 0.25 9.46

Moisture Content at 30°C

30 12.89 13.04 12.97 0.106 0.08 6.67

60 11.85 11.75 11.8 0.071 0.05 6.79

120 10.82 10.88 10.85 0.042 0.03 7.83

240 10.77 10.49 10.63 0.198 0.14 9.35

360 9.79 10.45 10.12 0.467 0.33 8.26

480 10.56 11.78 11.17 0.863 0.61 8.12

Moisture Content at 40°C

Page 111: Cassava starch as modified release excipient in selected

110

30 11.31 11.23 11.27 0.057 0.04 9.67

60 10.81 10.29 10.55 0.368 0.26 9.40

120 9.57 9.97 9.77 0.283 0.20 8.86

240 8.06 8.07 8.07 0.007 0.01 9.06

360 8.72 8.62 8.67 0.070 0.05 5.01

480 8.78 9.07 8.93 0.205 0.15 4.39

Moisture Content at 50°C

30 9.62 9.65 9.65 0.042 0.03 5.81

60 8.81 8.76 8.79 0.035 0.03 8.79

120 6.76 6.50 6.63 0.184 0.13 1.60

240 6.33 6.21 6.27 0.084 0.06 7.07

360 6.52 6.38 6.45 0.099 0.07 4.52

480 6.66 6.81 6.74 0.106 0.08 2.18

Table A.II: Karl-Fischer titration values for moisture content of the purchased Cassava starch

Purchased Starch

Time

(min)

Moisture (%H2O) %Weight

Loss Run 1 Run 2 Average SD %RSD

Reference Sample

0 11.95 11.73 11.84 0.156 0.11 7.89

Moisture Content at 25°C

30 12.28 11.81 12.05 0.332 0.24 4.47

60 12.61 12.51 12.56 0.071 0.05 10.19

Page 112: Cassava starch as modified release excipient in selected

111

120 12.1 12.37 12.24 0.191 0.14 7.44

240 12.59 12.42 12.51 0.120 0.09 7.87

360 12.39 11.62 12.01 0.544 0.39 10.64

480 11.64 11.82 11.73 0.127 0.09 6.61

Moisture Content at 30°C

30 11.63 11.75 11.69 0.085 0.06 10.10

60 11.58 11.39 11.49 0.134 0.10 9.94

120 11.2 10.63 10.92 0.403 0.29 8.79

240 10.14 9.97 10.06 0.120 0.09 3.43

360 10.19 10.18 10.19 0.007 0.01 8.79

480 10.69 10.74 10.72 0.035 0.03 3.88

Moisture Content at 40°C

30 10.13 9.82 9.98 0.219 0.11 4.49

60 9.46 9.25 9.36 0.148 0.07 3.91

120 8.26 8.29 8.28 0.021 0.01 8.47

240 9.06 8.51 8.79 0.389 0.19 10.06

360 8.02 8.01 8.02 0.007 0.00 3.86

480 8.36 8.26 8.31 0.070 0.04 8.86

Moisture Content at 50°C

30 8.91 8.87 8.89 0.028 0.02 4.97

60 7.72 7.71 7.72 0.007 0.01 8.19

120 7.21 7.04 7.13 0.120 0.09 3.65

240 6.29 6.81 6.55 0.368 0.26 2.92

Page 113: Cassava starch as modified release excipient in selected

112

360 6.06 6.68 6.37 0.438 0.31 7.15

480 6.84 7.13 6.99 0.205 0.15 4.25

MALVERN MASTERSIZER®

SIZE DISTRIBUTION

Particle Size Distribution

0.01 0.1 1 10 100 1000 10000

Particle Size (µm)

0

2

4

6

8

10

12

14

Volum

e (%

)

Cassava powder, 11 December 2014 11:36:16 AM

Particle Size Distribution

0.01 0.1 1 10 100 1000 10000

Particle Size (µm)

0

2

4

6

8

10

12

Volum

e (%

)

Cassava powder, 11 December 2014 11:53:39 AM

Particle Size Distribution

0.01 0.1 1 10 100 1000 10000

Particle Size (µm)

0

2

4

6

8

10

12

14

16

Volum

e (%

)

Formule 15, 11 December 2014 02:04:56 PM

Particle Size Distribution

0.01 0.1 1 10 100 1000 10000

Particle Size (µm)

0

2

4

6

8

10

12

14

16

18

Volum

e (%

)

Formule 11, 11 December 2014 01:39:54 PM

Page 114: Cassava starch as modified release excipient in selected

113

Particle Size Distribution

0.01 0.1 1 10 100 1000 10000

Particle Size (µm)

0

2

4

6

8

10

12

14

16 V

olum

e (%

)

Formule 21, 11 December 2014 02:08:14 PM

Particle Size Distribution

0.01 0.1 1 10 100 1000 10000

Particle Size (µm)

0

2

4

6

8

10

12

14

16

Volum

e (%

)

Formule 39, 11 December 2014 02:20:16 PM

Particle Size Distribution

0.01 0.1 1 10 100 1000 10000

Particle Size (µm)

0

2

4

6

8

10

12

14

16

Volum

e (%

)

Formule 46, 11 December 2014 02:35:02 PM

Particle Size Distribution

0.01 0.1 1 10 100 1000 10000

Particle Size (µm)

0

5

10

15

20

Volum

e (%

)

Formule 64, 11 December 2014 02:53:47 PM

Particle Size Distribution

0.01 0.1 1 10 100 1000 10000

Particle Size (µm)

0

5

10

15

20

Volum

e (%

)

Formule 74, 11 December 2014 03:04:24 PM

Figure A.I: Example graphs of size distribution graphs for starch powders and bead

formulations

Page 115: Cassava starch as modified release excipient in selected

Table A.III: Size distribution values starch powders and bead formulations

Donated starch Purchased starch C.G5.5.5 (Formula 15)

Run d(0.1) d(0.5) d(0.9) Run d(0.1) d(0.5) d(0.9) Run d(0.1) d(0.5) d(0.9)

1 7.57 14.09 24.97 1 7.52 15.68 27.56 1 11.92 809.98 1282.51

2 7.51 13.99 24.48 2 7.40 15.36 26.76 2 15.06 844.49 1292.73

3 7.53 14.02 24.56 3 7.27 15.10 26.19 3 22.25 800.28 1229.79

AVE 7.54 14.03 24.67 AVE 7.39 15.38 26.83 AVE 16.41 818.25 1268.35

SD 0.032 0.053 0.263 SD 0.125 0.293 0.688 SD 5.293 23.240 33.776

%RSD 0.43 0.38 1.07 %RSD 1.69 1.91 2.56 %RSD 32.26 2.84 2.66

M.G5.3.5 (Formula 11) M.G5.5.10 (Formula 21) M.G10.5.5 (Formula 39)

Run d(0.1) d(0.5) d(0.9) Run d(0.1) d(0.5) d(0.9) Run d(0.1) d(0.5) d(0.9)

1 130.48 915.14 1324.80 1 42.20 891.28 1353.65 1 625.00 974.89 1520.93

2 99.55 915.95 1326.34 2 33.04 909.48 1351.75 2 608.12 930.42 1303.36

3 80.79 939.39 1316.49 3 80.58 998.27 1461.19 3 583.63 955.66 1327.95

AVE 103.61 923.49 1322.54 AVE 51.94 933.01 1388.86 AVE 605.58 953.66 1384.08

SD 25.091 13.776 5.301 SD 25.223 57.246 62.647 SD 20.803 22.303 119.153

%RSD 24.22 1.49 0.40 %RSD 48.56 6.14 4.51 %RSD 3.44 2.34 8.61

Page 116: Cassava starch as modified release excipient in selected

M.G10.0.10 (Formula 46) M.G15.0.5 (Formula 64) M.G15.3.10 (Formula 74)

Run d(0.1) d(0.5) d(0.9) Run d(0.1) d(0.5) d(0.9) Run d(0.1) d(0.5) d(0.9)

1 685.42 1052.65 1579.03 1 635.24 875.93 1221.62 1 557.14 881.13 1236.32

2 665.75 1028.05 1559.55 2 635.91 888.56 1266.07 2 512.80 921.78 1295.57

3 649.22 1009.31 1540.53 3 673.76 920.55 1269.95 3 672.52 1013.07 1513.84

AVE 666.80 1030.00 1559.70 AVE 648.30 895.01 1252.55 AVE 580.82 938.66 1348.58

SD 18.123 21.7367 19.249 SD 22.048 22.998 26.857 SD 82.454 67.569 146.157

%RSD 2.72 2.11 1.23 %RSD 3.40 2.57 2.14 %RSD 14.20 7.20 10.84

Page 117: Cassava starch as modified release excipient in selected

116

ANNEXURE B

FLOW AND PHYSICAL PROPERTIES

Page 118: Cassava starch as modified release excipient in selected

FLOW PROPERTIES

Table B.I: Time and flow rate for Cassava starch powders and beads

Samples Mass

(g)

Time (s) Flow Rate (g.s-1)

1 2 3 Ave SD 1 2 3 Ave SD

Donated 100.14 No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

Purchased 101.07 No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

M.G5.3.5 99.59 22.00 20.00 21.00 21.00 1.000 4.53 4.98 4.74 4.75 0.226

M.G5.5.10 100.84 22.00 21.00 21.00 21.33 0.577 4.58 4.80 4.80 4.73 0.126

M.G10.5.5 99.72 22.00 21.00 22.00 21.67 0.577 4.53 4.75 4.53 4.60 0.125

M.G10.0.10 99.97 19.00 19.00 20.00 19.33 0.577 5.26 5.26 5.00 5.17 0.152

M.G15.0.5 100.02 26.00 26.00 27.00 26.33 0.577 3.85 3.85 3.70 3.80 0.082

M.G15.3.10 100.73 28.00 28.00 27.00 27.67 0.577 3.60 3.60 3.73 3.64 0.077

C.G5.5.5 100.10 20.00 20.00 21.00 20.33 0.577 5.01 5.01 4.77 4.93 0.138

Page 119: Cassava starch as modified release excipient in selected

Table B.II: Parameters relating to angle of repose, angle of repose and critical orifice diameter

Samples Mass

(g)

h (mm) r (mm)

1 2 3 Ave SD 1 2 3 Ave SD

Donated 100.14 No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

Purchased 101.07 No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

M.G5.3.5 99.59 3.00 3.10 2.90 3.00 0.100 5.95 6.15 5.85 5.98 0.153

M.G5.5.10 100.84 3.00 3.00 3.10 3.03 0.058 5.00 6.21 5.80 5.67 0.615

M.G10.5.5 99.72 3.10 2.70 2.90 2.90 0.200 6.05 6.00 6.00 6.02 0.029

M.G10.0.10 99.97 3.20 3.30 3.30 3.27 0.058 6.20 6.25 5.80 6.08 0.247

M.G15.0.5 100.02 3.10 3.50 3.30 3.30 0.200 5.95 5.60 5.75 5.77 0.176

M.G15.3.10 100.73 3.40 3.40 3.50 3.43 0.058 5.80 5.90 6.05 5.92 0.126

C.G5.5.5 100.10 2.90 3.00 3.00 2.97 0.058 6.00 5.50 6.20 6.02 0.361

Samples Mass

(g)

AoR (°) COD (mm)

1 2 3 Ave SD 1 2 3 Ave SD

Donated 100.14 No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

16.00 16.00 16.00 16.00 0.000

Purchased 101.07 No

Flow

No

Flow

No

Flow

No

Flow

No

Flow

16.00 16.00 16.00 16.00 0.000

M.G5.3.5 99.59 26.76 26.75 26.37 26.63 0.223 5.00 6.00 6.00 5.67 0.577

M.G5.5.10 100.84 30.96 25.78 28.12 28.29 2.594 6.00 6.00 6.00 6.00 0.000

M.G10.5.5 99.72 27.13 24.23 25.80 25.72 1.453 6.00 6.00 6.00 6.00 0.000

Page 120: Cassava starch as modified release excipient in selected

M.G10.0.10 99.97 27.30 27.83 29.64 28.26 1.226 6.00 7.00 7.00 6.67 0.577

M.G15.0.5 100.02 27.52 32.01 29.85 29.79 2.243 6.00 6.00 6.00 6.00 0.000

M.G15.3.10 100.73 30.38 29.95 30.05 30.13 0.223 6.00 6.00 6.00 6.00 0.000

C.G5.5.5 100.10 25.61 26.57 26.57 26.25 0.551 6.00 6.00 6.00 6.00 0.000

DENSITIES & COMPRESSIBILITY

Table B.III Volumes, densities and compressibility data for both starches and bead formulations

Bulk

Samples Mass

(g)

Volume (cm3) Density (g.cm-3)

1 2 3 Ave SD 1 2 3 Ave SD

Donated 100.14 168.00 166.00 164.00 166.00 2.000 0.60 0.60 0.61 0.60 0.007

Purchased 101.07 192.00 182.00 180.00 184.67 6.429 0.53 0.56 0.56 0.55 0.019

M.G5.3.5 99.59 138.00 124.00 122.00 128.00 8.718 0.72 0.80 0.82 0.78 0.051

M.G5.5.10 100.84 120.00 122.00 122.00 121.33 1.155 0.84 0.83 0.83 0.83 0.008

M.G10.5.5 99.72 126.00 126.00 124.00 125.33 1.155 0.79 0.79 0.80 0.80 0.007

M.G10.0.10 99.97 124.00 122.00 125.00 123.67 1.528 0.81 0.82 0.80 0.81 0.010

M.G15.0.5 100.02 120.00 121.00 121.00 120.67 0.577 0.83 0.83 0.83 0.83 0.004

M.G15.3.10 100.73 128.00 127.00 125.00 126.67 1.528 0.79 0.79 0.81 0.80 0.010

C.G5.5.5 100.10 121.00 122.00 123.00 122.00 1.000 0.83 0.82 0.81 0.82 0.007

Tapped

Samples Mass

(g)

Volume (cm3) Density (g.cm-3)

1 2 3 Ave SD 1 2 3 Ave SD

Donated 100.14 124.00 122.00 118.00 121.33 3.055 0.81 0.82 0.85 0.83 0.021

Purchased 101.07 130.00 124.00 120.00 124.67 5.033 0.78 0.82 0.84 0.81 0.033

M.G5.3.5 99.59 110.00 110.00 112.00 110.67 1.155 0.91 0.91 0.89 0.90 0.009

M.G5.5.10 100.84 116.00 114.00 114.00 114.67 1.155 0.87 0.88 0.88 0.88 0.009

Page 121: Cassava starch as modified release excipient in selected

M.G10.5.5 99.72 114.00 114.00 114.00 114.00 0.000 0.87 0.87 0.87 0.87 0.000

M.G10.0.10 99.97 111.00 110.00 111.00 110.67 0.577 0.90 0.91 0.90 0.90 0.005

M.G15.0.5 100.02 115.00 115.00 116.00 115.33 0.577 0.87 0.87 0.86 0.87 0.004

M.G15.3.10 100.73 116.00 116.00 116.00 116.00 0.000 0.87 0.87 0.87 0.87 0.000

C.G5.5.5 100.10 114.00 114.00 115.00 114.33 0.577 0.88 0.88 0.87 0.88 0.004

Packed

Samples Mass

(g)

Volume (cm3) Packed Density (g.cm-3)

1 2 3 Ave SD 1 2 3 Ave SD

Donated 100.14 44.00 44.00 46.00 44.67 1.155 2.28 2.28 2.18 2.24 0.057

Purchased 101.07 62.00 58.00 60.00 60.00 2.000 1.63 1.74 1.68 1.69 0.056

M.G5.3.5 99.59 28.00 14.00 10.00 17.33 9.452 3.56 7.11 9.96 6.88 3.208

M.G5.5.10 100.84 4.00 8.00 8.00 6.67 2.309 25.21 12.61 12.61 16.81 7.278

M.G10.5.5 99.72 12.00 12.00 10.00 11.33 1.155 8.31 8.31 9.97 8.86 0.960

M.G10.0.10 99.97 13.00 12.00 14.00 13.00 1.000 7.69 8.33 7.14 7.72 0.596

M.G15.0.5 100.02 5.00 6.00 5.00 5.33 0.577 20.00 16.67 20.00 18.89 1.925

M.G15.3.10 100.73 12.00 11.00 9.00 10.67 1.528 8.39 9.16 11.19 9.58 1.446

C.G5.5.5 100.10 7.00 8.00 8.00 7.67 0.577 14.30 12.51 12.51 13.11 1.032

Compressibility

Samples Mass

(g)

Carr's index Hausner Ration

1 2 3 Ave SD 1 2 3 Ave SD

Donated 100.14 0.26 0.27 0.28 0.27 0.010 1.35 1.36 1.39 1.37 0.019

Purchased 101.07 0.32 0.32 0.33 0.32 0.008 1.48 1.47 1.50 1.48 0.017

M.G5.3.5 99.59 0.20 0.11 0.08 0.13 0.063 1.25 1.13 1.09 1.16 0.087

M.G5.5.10 100.84 0.06 0.07 0.07 0.06 0.004 1.06 1.07 1.07 1.07 0.005

M.G10.5.5 99.72 0.03 0.07 0.07 0.05 0.019 1.03 1.07 1.07 1.06 0.021

M.G10.0.10 99.97 0.10 0.10 0.08 0.09 0.008 1.11 1.11 1.09 1.10 0.010

M.G15.0.5 100.02 0.10 0.10 0.11 0.11 0.007 1.12 1.11 1.13 1.12 0.009

M.G15.3.10 100.73 0.04 0.05 0.04 0.04 0.005 1.04 1.05 1.04 1.05 0.005

C.G5.5.5 100.10 0.09 0.09 0.07 0.08 0.011 1.10 1.09 1.08 1.09 0.013

Page 122: Cassava starch as modified release excipient in selected

121

SWELLING AND EROSION

Table B.IV: Swelling and erosion data for Avicel® bead formulations

Time

(min) Parameter

M.G5.3.5

Run 1 Run 2 Run 3 Average SD

Initial mass 251.50 249.00 249.50 250.00 0.500

30

Mass 1429.76 1424.00 1416.76 1423.51 4.045

%Swelling 102.71 102.69 102.68 102.69 0.009

%Erosion 177.37 175.20 173.94 175.50 0.829

60

Mass 1230.20 1205.50 1198.80 1211.50 6.353

%Swelling 102.25 102.19 102.18 102.20 0.015

%Erosion 176.58 174.34 173.09 174.67 0.834

90

Mass 1269.80 1218.80 1214.30 1234.30 10.492

%Swelling 102.34 102.22 102.21 102.26 0.024

%Erosion 176.74 174.39 173.15 174.76 0.844

120

Mass 1259.60 1181.40 1178.70 1206.57 15.369

%Swelling 102.31 102.14 102.13 102.19 0.035

%Erosion 176.70 174.25 173.01 174.65 0.854

180

Mass 1923.80 1873.10 1857.90 1884.93 13.552

%Swelling 103.84 103.72 103.69 103.75 0.031

%Erosion 179.33 176.96 175.66 177.32 0.874

240

Mass 1929.70 1920.90 1910.30 1920.30 5.954

%Swelling 103.85 103.83 103.81 103.83 0.014

%Erosion 179.36 177.15 175.86 177.45 0.846

360

Mass 1832.40 1839.80 1819.30 1830.50 10.265

%Swelling 103.63 103.65 103.60 103.63 0.024

%Erosion 178.97 176.83 175.51 177.10 0.853

480

Mass 1839.20 1766.80 1739.20 1781.73 21.579

%Swelling 103.65 103.48 103.42 103.51 0.050

%Erosion 179.00 176.54 175.19 176.91 0.904

600 Mass 2010.10 1926.70 1905.70 1947.50 20.900

Page 123: Cassava starch as modified release excipient in selected

122

%Swelling 104.04 103.85 103.80 103.89 0.048

%Erosion 179.67 177.17 175.84 177.56 0.901

720

Mass 1883.60 1812.20 1801.70 1832.50 15.658

%Swelling 103.75 103.58 103.56 103.63 0.036

%Erosion 179.17 176.72 175.44 177.12 0.875

Dried Mass 172.70 170.60 169.40 170.90 0.794

Time (min)

Parameter M.G5.5.10

Run 1 Run 2 Run 3 Average SD

Initial mass 250.30 249.70 251.20 250.40 0.751

30

Mass 1367.50 1301.80 1284.30 1317.87 16.788

%Swelling 102.58 102.43 102.39 102.47 0.039

%Erosion 175.63 173.93 175.60 175.05 0.852

60

Mass 1421.70 1404.10 1348.80 1391.53 28.989

%Swelling 102.71 102.67 102.54 102.64 0.067

%Erosion 175.84 174.33 175.86 175.34 0.776

90

Mass 1446.30 1375.30 1353.30 1391.63 19.236

%Swelling 102.77 102.60 102.55 102.64 0.045

%Erosion 175.94 174.22 175.88 175.34 0.845

120

Mass 1621.20 1564.30 1515.60 1567.03 28.938

%Swelling 103.17 103.04 102.93 103.05 0.067

%Erosion 176.63 174.96 176.52 176.04 0.797

180

Mass 1711.20 1663.00 1628.30 1667.50 21.451

%Swelling 103.38 103.27 103.19 103.28 0.050

%Erosion 176.99 175.35 176.97 176.43 0.824

240

Mass 1541.40 1503.40 1495.10 1513.30 9.112

%Swelling 102.99 102.90 102.88 102.92 0.021

%Erosion 176.31 174.72 176.44 175.83 0.869

360

Mass 1465.70 1469.00 1460.40 1465.03 4.304

%Swelling 102.81 102.82 102.80 102.81 0.010

%Erosion 176.01 174.59 176.30 175.63 0.863

480

Mass 1642.20 1573.00 1500.20 1571.80 41.689

%Swelling 103.22 103.06 102.89 103.06 0.096

%Erosion 176.71 175.00 176.46 176.06 0.755

600

Mass 1774.60 1741.00 1743.70 1753.10 6.352

%Swelling 103.53 103.45 103.46 103.48 0.014

%Erosion 177.24 175.66 177.42 176.77 0.894

720 Mass 1562.00 1525.30 1501.30 1529.53 15.226

%Swelling 103.04 102.95 102.89 102.96 0.033

Page 124: Cassava starch as modified release excipient in selected

123

%Erosion 176.40 174.81 176.46 175.89 0.840

Dried Mass 171.20 169.80 171.50 170.83 0.857

Time (min)

Parameter M.G10.5.5

Run 1 Run 2 Run 3 Average SD

Initial mass 250.40 251.10 250.30 250.60 0.404

30

Mass 1257.10 1251.00 1222.60 1243.57 14.728

%Swelling 102.29 102.27 102.21 102.26 0.033

%Erosion 174.30 178.06 175.80 176.05 1.237

60

Mass 1051.80 1039.50 1019.60 1036.97 10.832

%Swelling 101.82 101.79 101.75 101.79 0.025

%Erosion 173.50 177.22 175.01 175.24 1.215

90

Mass 1247.40 1227.40 1223.80 1232.87 4.565

%Swelling 102.27 102.22 102.21 102.23 0.010

%Erosion 174.26 177.96 175.81 176.01 1.191

120

Mass 1234.10 1213.10 1214.40 1220.53 3.970

%Swelling 102.24 102.19 102.19 102.21 0.009

%Erosion 174.21 177.91 175.77 175.96 1.183

180

Mass 724.70 675.90 654.40 685.00 15.713

%Swelling 101.08 100.97 100.92 100.99 0.036

%Erosion 172.24 175.78 173.58 173.87 1.197

240

Mass 664.70 688.00 667.70 673.47 10.461

%Swelling 100.94 100.99 100.95 100.96 0.024

%Erosion 172.00 175.83 173.63 173.82 1.218

360

Mass 341.40 352.90 354.70 349.67 2.550

%Swelling 100.21 100.23 100.24 100.23 0.006

%Erosion 170.75 174.50 172.41 172.56 1.170

480

Mass 637.10 588.60 571.60 599.10 13.877

%Swelling 100.88 100.77 100.73 100.79 0.032

%Erosion 171.90 175.44 173.26 173.53 1.188

600

Mass 765.70 737.30 714.50 739.17 13.734

%Swelling 101.17 101.11 101.05 101.11 0.031

%Erosion 172.40 176.03 173.82 174.08 1.207

720

Mass 555.10 552.50 542.90 550.17 5.007

%Swelling 100.69 100.69 100.66 100.68 0.011

%Erosion 171.58 175.29 173.14 173.34 1.189

Dried Mass 170.40 174.10 172.00 172.17 1.167

Time (min)

Parameter M.G10.0.10

Run 1 Run 2 Run 3 Average SD

Initial mass 250.10 251.70 251.20 251.00 0.361

30 Mass 1303.40 1212.70 1202.60 1239.57 19.107

Page 125: Cassava starch as modified release excipient in selected

124

%Swelling 102.49 102.28 102.25 102.34 0.045

%Erosion 174.34 177.04 174.54 175.31 1.279

60

Mass 1296.30 1273.50 1216.80 1262.20 30.010

%Swelling 102.47 102.42 102.29 102.39 0.071

%Erosion 174.31 177.29 174.60 175.40 1.380

90

Mass 1409.20 1365.10 1341.40 1371.90 16.011

%Swelling 102.74 102.64 102.58 102.65 0.038

%Erosion 174.76 177.66 175.10 175.84 1.317

120

Mass 1459.10 1402.80 1417.80 1426.57 12.019

%Swelling 102.86 102.73 102.76 102.78 0.028

%Erosion 174.96 177.82 175.41 176.06 1.244

180

Mass 898.50 865.50 874.70 879.57 7.144

%Swelling 101.53 101.46 101.48 101.49 0.017

%Erosion 172.71 175.62 173.22 173.85 1.243

240

Mass 1008.60 980.60 983.90 991.03 5.333

%Swelling 101.79 101.73 101.74 101.75 0.013

%Erosion 173.15 176.09 173.66 174.30 1.258

360

Mass 1398.70 1460.40 1457.60 1438.90 11.689

%Swelling 102.72 102.86 102.86 102.81 0.028

%Erosion 174.72 178.05 175.57 176.11 1.304

480

Mass 891.90 882.80 865.90 880.20 9.100

%Swelling 101.52 101.50 101.46 101.49 0.022

%Erosion 172.68 175.69 173.18 173.85 1.297

600

Mass 961.80 916.60 820.10 899.50 51.493

%Swelling 101.68 101.58 101.35 101.54 0.122

%Erosion 172.96 175.83 173.00 173.93 1.441

720

Mass 1168.20 1151.60 1121.50 1147.10 16.236

%Swelling 102.17 102.13 102.06 102.12 0.038

%Erosion 173.79 176.79 174.22 174.93 1.328

Dried Mass 170.10 173.10 170.70 171.30 1.249

Time (min)

Parameter M.G15.0.5

Run 1 Run 2 Run 3 Average SD

Initial mass 250.40 251.20 251.10 250.90 0.153

30

Mass 1164.10 1143.20 1085.10 1130.80 30.599

%Swelling 102.14 102.10 101.96 102.07 0.072

%Erosion 175.28 169.48 175.27 173.34 2.950

60

Mass 1166.20 1114.90 1083.90 1121.67 20.137

%Swelling 102.15 102.03 101.96 102.04 0.047

%Erosion 175.29 169.37 175.26 173.31 3.004

90 Mass 1069.50 1042.90 1038.20 1050.20 6.047

Page 126: Cassava starch as modified release excipient in selected

125

%Swelling 101.92 101.86 101.85 101.88 0.014

%Erosion 174.90 169.09 175.08 173.02 3.046

120

Mass 1137.90 1180.40 1151.60 1156.63 15.382

%Swelling 102.08 102.18 102.11 102.13 0.036

%Erosion 175.17 169.62 175.54 173.44 3.000

180

Mass 1447.80 1411.90 1382.40 1414.03 17.680

%Swelling 102.81 102.73 102.66 102.73 0.041

%Erosion 176.42 170.52 176.47 174.47 3.025

240

Mass 1259.60 1257.00 1242.00 1252.87 7.748

%Swelling 102.37 102.36 102.33 102.35 0.018

%Erosion 175.66 169.92 175.90 173.83 3.038

360

Mass 1368.80 1359.70 1341.70 1356.73 9.651

%Swelling 102.62 102.60 102.56 102.60 0.023

%Erosion 176.10 170.32 176.30 174.24 3.040

480

Mass 1426.10 1354.60 1325.60 1368.77 22.004

%Swelling 102.76 102.59 102.52 102.62 0.052

%Erosion 176.33 170.30 176.24 174.29 3.028

600

Mass 1502.50 1466.20 1433.50 1467.40 19.235

%Swelling 102.94 102.85 102.78 102.86 0.045

%Erosion 176.64 170.73 176.67 174.68 3.023

720

Mass 1290.60 1265.90 1263.20 1273.23 5.192

%Swelling 102.44 102.38 102.38 102.40 0.012

%Erosion 175.79 169.95 175.99 173.91 3.065

Dried Mass 171.60 166.00 171.90 169.83 2.994

Time (min)

Parameter M.G15.0.5

Run 1 Run 2 Run 3 Average SD

Initial mass 250.20 250.90 251.30 250.80 0.265

30

Mass 1323.30 1290.90 1304.30 1306.17 8.328

%Swelling 102.51 102.44 102.47 102.47 0.020

%Erosion 175.50 176.40 175.63 175.84 0.395

60

Mass 1112.00 1126.50 1090.60 1109.70 17.962

%Swelling 102.02 102.05 101.97 102.01 0.042

%Erosion 174.65 175.73 174.77 175.05 0.494

90

Mass 1065.40 1062.20 1073.80 1067.13 5.822

%Swelling 101.91 101.90 101.93 101.91 0.014

%Erosion 174.47 175.47 174.70 174.88 0.402

120

Mass 1216.90 1171.00 1174.10 1187.33 8.675

%Swelling 102.26 102.16 102.16 102.19 0.020

%Erosion 175.08 175.91 175.11 175.37 0.411

180 Mass 1377.60 1367.60 1359.60 1368.27 4.823

Page 127: Cassava starch as modified release excipient in selected

126

%Swelling 102.64 102.62 102.60 102.62 0.011

%Erosion 175.72 176.71 175.85 176.09 0.440

240

Mass 1217.80 1229.00 1198.10 1214.97 15.472

%Swelling 102.27 102.29 102.22 102.26 0.036

%Erosion 175.08 176.15 175.20 175.48 0.485

360

Mass 1124.10 1109.00 1105.40 1112.83 3.717

%Swelling 102.05 102.01 102.00 102.02 0.009

%Erosion 174.70 175.66 174.83 175.07 0.428

480

Mass 1340.30 1294.60 1283.50 1306.13 11.317

%Swelling 102.55 102.45 102.42 102.47 0.027

%Erosion 175.57 176.41 175.55 175.84 0.439

600

Mass 1416.40 1356.70 1321.80 1364.97 22.912

%Swelling 102.73 102.59 102.51 102.61 0.054

%Erosion 175.88 176.66 175.70 176.08 0.484

720

Mass 1159.10 1189.70 1174.10 1174.30 8.949

%Swelling 102.13 102.20 102.16 102.16 0.021

%Erosion 174.84 175.99 175.11 175.31 0.460

Dried Mass 171.20 172.20 171.40 171.60 0.416

Table B.V: Friability parameters and data

Sample Parameters 1 2 3 Ave SD

M.G5.3.5 W0 3.00 3.00 3.01 3.00 0.005

W1 2.84 3.01 3.01 2.96 0.100

%Friability 5.33 0.00 0.00 1.78 3.079

M.G5.5.10 W0 3.00 3.00 3.00 3.00 0.001

W1 3.00 2.99 3.01 3.00 0.008

%Friability 0.00 0.37 0.00 0.12 0.212

M.G10.5.5 W0 3.00 3.00 3.00 3.00 0.001

W1 3.01 3.01 3.01 3.01 0.003

%Friability 0.00 0.00 0.00 0.00 0.000

M.G10.0.10 W0 3.00 3.00 3.00 3.00 0.001

W1 2.98 2.99 2.98 2.98 0.005

%Friability 0.63 0.47 0.87 0.66 0.201

M.G15.0.5 W0 3.00 3.00 3.01 3.00 0.002

W1 3.01 3.01 3.02 3.01 0.005

%Friability 0.00 0.00 0.00 0.00 0.000

M.G15.3.10 W0 3.00 3.00 3.00 3.00 0.002

W1 3.01 3.04 3.01 3.02 0.015

%Friability 0.00 0.00 0.00 0.00 0.000

Page 128: Cassava starch as modified release excipient in selected

127

C.G.5.5.5 W0 3.00 3.00 3.00 3.00 0.001

W1 2.91 2.97 2.95 2.94 0.027

%Friability 2.87 1.10 1.90 1.96 0.885

Table B.VI Disintegration times

Samples

Disintegration time (s)

Ave SD Vessel 1

Vessel 2

Vessel 3

Vessel 4

Vessel 5

Vessel 6

M.G5.3.5 241.80 247.80 259.20 240.60 252.00 249.00 248.40 6.852

M.G5.5.10 300.00 258.00 313.80 420.60 378.00 480.00 358.40 83.090

M.G10.5.5 420.00 510.00 486.00 498.00 540.00 486.00 490.00 39.739

M.G10.0.10 275.40 336.00 288.00 258.00 330.00 354.00 306.90 38.313

M.G15.0.5 600.00 606.00 564.00 570.00 330.00 384.00 509.00 120.085

M.G15.3.10 300.60 270.00 510.00 396.00 450.00 330.00 376.10 92.595

C.G5.5.5 210.00 270.00 330.00 300.00 294.00 198.00 267.00 52.547

Page 129: Cassava starch as modified release excipient in selected

128

ANNEXURE C

DISSOLUTION STUDIES

Page 130: Cassava starch as modified release excipient in selected

129

LINEARITY AND VALIDATION

Table C.I: Linearity and validation data for gliclazide in acidic medium

Medium Acidic Day 1 Run 1 Intraday

Standard Gliclazide (µg.ml-1)

Average Absorbance

SD %RSD Mass (mg)

25.1

1 2.01 0.06 0.00 0.08

Regression 2 5.02 0.19 0.00 0.05

3 10.04 0.39 0.00 0.04

4 20.08 0.74 0.00 0.03 m 0.04

5 30.12 1.13 0.06 4.01 c -0.00

6 40.16 1.49 0.0 0.06 r 0.9998

Medium Acidic Day 1 Run 2 Intraday

Standard Gliclazide (µg.ml-1)

Average Absorbance

SD %RSD Mass (mg)

25.1

1 2 0.54 0.00 0.04

Regression 2 5 0.095 0.00 0.04

3 10 0.46 0.00 0.02

4 20 0.83 0.00 0.00 m 0.19

5 30 1.20 0.00 0.01 c -0.10

6 40 1.59 0.00 0.10 r 0.9999

Medium Acidic Day 1 Run 3 Intraday

Standard Gliclazide (µg.ml-1)

Average Absorbance

SD %RSD Mass (mg)

25.1

1 2.01 0.06 0.01 0.10

Regression 2 5.02 0.10 0.00 0.00

3 10.04 0.43 0.12 0.20

4 20.08 0.81 0.46 0.00 m 0.18

5 30.12 1.23 0.00 2.00 c -0.10

6 40.16 1.50 1.29 1.07 r 0.9997

Medium Acidic Day 2 Run 1 Interday

Standard Gliclazide (µg.ml-1)

Average Absorbance

SD %RSD Mass (mg)

25.1

1 2.01 0.08 0.01 12.63

Regression 2 5.02 0.21 0.00 0.19

3 10.04 0.43 0.01 1.06

4 20.08 0.82 0.03 2.41 m 0.04

5 30.12 1.20 0.21 12.40 c 0.02

6 40.16 1.59 0.00 0.21 r 0.9997

Medium Acidic Day 3 Run 1 Interday

Page 131: Cassava starch as modified release excipient in selected

130

Standard Gliclazide (µg.ml-1)

Average Absorbance

SD %RSD Mass (mg)

25.1

1 2.01 -0.04 0.00 -0.19

Regression 2 5.02 0.08 0.00 0.09

3 10.04 0.30 0.00 0.22

4 20.08 0.67 0.00 0.26 m 0.04

5 30.12 1.06 0.00 0.01 c -0.11

6 40.16 1.46 0.00 0.11 r 0.9999

Table C.II Linearity and validation data for gliclazide in alkaline medium

Medium Alkaline Day 1 Run 1 Intraday

Standard Gliclazide (µg.ml-1)

Average Absorbance

SD %RSD Mass (mg)

25

1 2 0.00 0.00 7.19

Regression 2 5 0.15 0.00 0.33

3 10 0.32 0.01 4.29

4 20 0.70 0.00 0.22 m 0.03

5 30 1.07 0.00 0.04 c -0.06

6 40 1.49 0.00 0.05 r 0.9995

Medium Alkaline Day 1 Run 2 Intraday

Standard Gliclazide (µg.ml-1)

Average Absorbance

SD %RSD Mass (mg)

25

1 2 0.01 0.00 0.01

Regression 2 5 0.23 0.00 0.09

3 10 0.33 0.00 2.70

4 20 0.59 0.00 0.00 m 0.19

5 30 1.10 0.02 0.01 c -0.30

6 40 1.53 0.01 0.00 r 0.992

Medium Alkaline Day 1 Run 3 Intraday

Standard Gliclazide (µg.ml-1)

Average Absorbance

SD %RSD Mass (mg)

25.1

1 2.01 0.18 0.00 0.03

Regression 2 5.02 0.24 0.00 0.30

3 10.04 0.33 0.00 0.87

4 20.08 0.77 1,00 0.99 m 0.1532

5 30,12 1.01 0.20 0.01 c -0.2698

6 40.16 1.40 0.00 0.00 r 0.9997

Medium Alkaline Day 2 Run 1 Interday

Standard Gliclazide (µg.ml-1)

Average Absorbance

SD %RSD Mass (mg)

24.9

Page 132: Cassava starch as modified release excipient in selected

131

1 1.992 0.17 0.00 0.33

Regression 2 4.98 0.27 0.00 0.04

3 9.96 0.43 0.00 0.05

4 19.92 0.77 0.00 0.05 m 0.03

5 29.88 1.08 0.01 0.49 c 0.10

6 39.84 1.46 0.00 0.15 r 0.9996

Medium Alkaline Day 3 Run 1 Interday

Standard Gliclazide (µg.ml-1)

Average Absorbance

SD %RSD Mass (mg)

25

1 2 0.00 0.00 3.30

Regression 2 5 0.11 0.00 0.33

3 10 0.33 0.00 0.04

4 20 0.69 0.00 0.01 m 0.04

5 30 1.07 0.01 0.50 c -0.06

6 40 1.42 0.15 7.30 r 0.9997

Page 133: Cassava starch as modified release excipient in selected

132

DISSOLUTION DATA

Table C.III: Dissolution data for bead formulations and Diamicron®

Time (min)

M.G5.3.5 M.G5.5.10 M.G10.5.5 M.G10.0.10

Ave SD %RSD Ave SD %RSD Ave SD %RSD Ave SD

0 0.00 0.000 0 0.00 0.000 0.00 0 0 0 0 0

2.5 3.55 1.314 36.99 3.07 0.263 8.58 19.97 1.074 5.38 5.93 0.222

5.0 6.11 0.933 15.28 4.80 0.771 16.06 21.60 0.659 3.05 7.54 0.948

7.5 8.86 0.889 10.03 7.22 1.639 22.71 23.44 0.825 3.52 9.86 0.766

15 11.8

8 2.333 19.63 13.62 2.828 20.76 26.80 3.898 14.54 11.07 0.897

30 16.8

4 2.983 17.71 26.71 9.634 36.07 31.70 3.947 12.45 16.56 1.104

60 20.3

8 4.393 21.56 42.06 2.831 6.73 37.33 1.876 5.03 19.93 1.438

90 26.5

3 3.096 11.67 48.04 2.457 5.11 45.88 0.979 2.13 26.48 1.964

120 30.5

8 1.891 6.18 55.03 5.574 10.13 51.90 3.122 6.02 34.22 0.619

180 32.1

9 2.223 6.91 59.66 3.082 5.17 54.96 3.104 5.65 39.62 3.415

240 39.7

3 2.516 6.33 63.26 3.535 5.59 64.90 4.356 6.71 44.16 1.413

360 44.4

1 2.899 6.53 68.63 2.061 3.00 66.53 2.965 4.46 69.14 5.569

480 59.0

4 10.281 17.41 84.77 9.409 11.10 76.50

11.73

0 15.33 81.41 1.999

600 87.0

2 7.824 8.99 94.29 2.784 2.95 90.93 6.453 7.10 87.17 0.586

720 92.8

1 4.433 4.78 97.55 2.058 2.11 98.86 0.424 0.43 95.96 1.517

735 100.00

0.000 0.00 100.0

0 0.000 0.00 100.00 0.000 0.00 100.00 0.000

Time (min)

M.G15.0.5 M.G15.3.10 C.G5.5.5 Diamicron®

Ave SD %RSD Ave SD %RSD Ave SD %RSD Ave SD

0 0 0 0 0 0 0 0 0 0 0 0

2.5 11.33 2.061 18.19 2.92 0.379 12.97 32.63 9.765 29.93 11.92 4.847

Page 134: Cassava starch as modified release excipient in selected

133

5.0 13.09 2.118 16.18 4.89 1.356 27.73 39.57 7.155 18.08 12.54 1.311

7.5 15.27 1.481 9.70 6.45 0.622 9.65 44.57 7.746 17.38 19.43 4.618

15 16.10 1.376 8.55 8.95 2.372 26.49 60.44 11.27

7 18.66 22.62 2.768

30 18.89 1.002 5.30 14.65 4.239 28.94 63.88 7.676 12.02 25.29 1.679

60 23.81 1.320 5.54 22.17 1.648 7.43 66.62 8.967 13.46 28.35 2.376

90 27.86 0.806 2.89 31.67 0.602 1.90 69.37 7.784 11.22 36.91 7.244

120 32.51 2.732 8.40 35.95 4.587 12.76 70.87 8.215 11.59 37.62 1.353

180 34.66 4.384 12.65 43.13 3.708 8.60 74.67 8.698 11.65 45.51 4.353

240 42.60 3.490 8.19 65.77 3.910 5.94 78.94 9.059 11.48 74.64 0.857

360 56.30 5.872 10.43 73.23 6.101 8.33 81.85 8.301 10.14 77.18 2.306

480 67.27 4.423 6.57 82.71 3.994 4.83 88.55 8.407 9.49 83.48 4.250

600 86.71 8.099 9.34 84.54 7.178 8.49 93.03 3.613 3.88 88.70 6.583

720 92.46 5.926 6.41 92.70 5.834 6.29 94.64 3.849 4.07 95.07 1.538

735 100.0

0 0.000 0.00

100.0

0 0.000 0.00 100.00 0.000 0.00 100.00 0.000